Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Br J Cancer ; 106(3): 496-507, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22240799

RESUMO

BACKGROUND: There are still no effective treatments for superficial bladder cancer (SBC)/non-muscle invasive bladder cancer. Following treatment, 20% of patients still develop metastatic disease. Superficial bladder cancer is often multifocal, has high recurrences after surgical resection and recurs after intravesical live Bacillus Calmette-Guérin. Oncovex(GALV/CD), an oncolytic herpes simplex virus-1, has shown enhanced local tumour control by combining oncolysis with the expression of a highly potent pro-drug activating gene and the fusogenic glycoprotein. METHODS: In vitro fusion/prodrug/apoptotic cell-based assays. In vivo orthotopic bladder tumour model, visualised by computed microtomography. RESULTS: Treatment of seven human bladder carcinoma cell lines with the virus resulted in tumour cell killing through oncolysis, pro-drug activation and glycoprotein fusion. Oncovex(GALV/CD) and mitomycin C showed a synergistic effect, whereas the co-administration with cisplatin or gemcitabine showed an antagonistic effect in vitro. Transitional cell cancer (TCC) cells follow an apoptotic cell death pathway after infection with Oncovex(GALV/CD) with or without 5-FC. In vivo results showed that intravesical treatment with Oncovex(GALV/CD) + prodrug (5-FC) reduced the average tumour volume by over 95% compared with controls. DISCUSSION: Our in vitro and in vivo results indicate that Oncovex(GALV/CD) can improve local tumour control within the bladder, and potentially alter its natural history.


Assuntos
Carcinoma de Células de Transição/terapia , Glicoproteínas/uso terapêutico , Recidiva Local de Neoplasia/terapia , Terapia Viral Oncolítica , Pró-Fármacos/uso terapêutico , Neoplasias da Bexiga Urinária/terapia , Administração Intravesical , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Cricetinae , Modelos Animais de Doenças , Feminino , Fluoruracila/farmacologia , Glicoproteínas/farmacologia , Herpesvirus Humano 1/genética , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Recidiva Local de Neoplasia/patologia , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacologia , Ratos , Ratos Endogâmicos F344 , Neoplasias da Bexiga Urinária/patologia
3.
Gene Ther ; 18(11): 1063-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21562589

RESUMO

Hepatocyte growth factor (HGF) promotes regeneration of the central nervous system, but its effects on the peripheral nervous system remain unclear. This study was conducted to elucidate the effect of HGF on regeneration of the murine facial nerve after crush injury. To do so, a replication-defective herpes simplex virus vector that incorporated HGF was prepared (HSV-HGF). The main trunk of the facial nerve was compressed by mosquito hemostats, and HSV-HGF, control vector or medium was then applied to the compressed nerve. We found that mice in the HGF group required significantly fewer days for complete recovery from nerve compression. Furthermore, the amplitude of the evoked buccinator muscle compound action potential increased following HSV-HGF application. HGF expression in and around the compressed nerve was demonstrated by enzyme-linked immunoassay and immunohistochemistry. In addition, HSV-HGF introduction around the damaged nerve significantly accelerated recovery of function of the facial nerve. These data suggest a possible role of HGF in promoting facial nerve regeneration after nerve damage. Furthermore, this viral delivery method may be applied clinically for many types of severe facial palsy during facial nerve decompression surgery.


Assuntos
Traumatismos do Nervo Facial/terapia , Terapia Genética/métodos , Fator de Crescimento de Hepatócito/genética , Regeneração Nervosa/efeitos dos fármacos , Simplexvirus/genética , Animais , Nervo Facial/fisiologia , Vetores Genéticos , Camundongos , Síndromes de Compressão Nervosa/terapia , Regeneração Nervosa/genética
4.
Restor Neurol Neurosci ; 26(6): 509-20, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19096139

RESUMO

PURPOSE: The primary motor pathway, the corticospinal tract, is a major target for spinal cord regeneration studies. One way of improving the regeneration of corticospinal axons is to introduce regeneration-associated genes into cortical motor neurons using viral vector delivery. METHODS: We used an engineered Herpes Simplex virus (HSV1) with the EF1alpha promoter encoding either LacZ or GFP to transduce cortical neurons through retrograde transport following the injection of vector into adult rat striatum or spinal cord. After three-days to one-month post-injection, sections of brain and spinal cord were viewed with fluorescence microscopy or processed for LacZ histochemistry. RESULTS: Many layer V motor cortical neurons were transduced following striatal injections. These were not corticospinal neurons as they were not fluorogold-labelled following tracer injection into spinal cord. Corticospinal neurons in both hemispheres were, however, transduced following direct vector injections into the dorsal column of spinal cord, yielding 250-400 transduced corticospinal neurons per animal. No non-pyramidal neurons or thalamic neurons were transduced by spinal injections. CONCLUSIONS: Therefore, this HSV1.EF1alpha vector is highly effective for the transduction of corticospinal neurons without direct injection into the brain and could be used to introduce regeneration-relevant genes into these neurons with the aim of regenerating the corticospinal tract.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Células Piramidais/fisiologia , Medula Espinal/fisiologia , Transdução Genética/métodos , Animais , Contagem de Células/métodos , Efrina-A2/genética , Efrina-A2/metabolismo , Feminino , Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Masculino , Tratos Piramidais/fisiologia , Ratos , Ratos Endogâmicos Lew , Estilbamidinas/metabolismo , beta-Galactosidase/genética
5.
J Gene Med ; 9(2): 99-106, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17256802

RESUMO

BACKGROUND: Tumour necrosis factor alpha (TNFalpha) therapy is a promising anti-cancer treatment when combined with radiotherapy due to its potent radio sensitising effects, but systemic toxicity has limited its clinical use. Previously, non-replicative adenovirus vectors have been used to deliver TNFalpha directly to the tumour, including under the control of a radiation sensitive promoter. Here, we have used an ICP34.5 deleted, oncolytic herpes simplex virus (HSV) for delivery to increase expression levels and spread through the tumour, and the use of the US11 true late HSV promoter to limit expression to where the virus replicates, i.e. selectively in tumour tissue. METHODS: TNFalpha expression under the CMV or US11 promoter was compared on cell lines CT26, BHK and Fadu. To further compare the activities of the promoters, expression of human TNFalpha was analysed in the presence and absence of acyclovir--an inhibitor of viral DNA replication and on HSV/ICP34.5- non-permissive cell line 3T6. The in vivo efficacy and toxicity of TNFalpha viruses were compared using A20 double flank tumour model in Balb/C mice and Fadu tumour model in nude mice. RESULTS: The results demonstrated that the US11 promoter significantly reduced and delayed TNFalpha expression as compared to use of the CMV promoter, especially in non-permissive cells or in the presence of acyclovir. Despite the reduced and more selective expression levels, US11 driven TNFalpha showed improved anti-tumour effects compared to CMV driven TNFalpha, and without the toxic side effects. CONCLUSIONS: This approach is therefore beneficial in increasing localised TNFalpha expression as compared to the use of non-replicative approaches, and combines the effects of TNFalpha with oncolytic virus replication which is expected to further enhance the efficacy of radiotherapy in a combined treatment approach.


Assuntos
Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Simplexvirus/genética , Fator de Necrose Tumoral alfa/genética , Animais , Cricetinae , Citomegalovirus/genética , Citomegalovirus/metabolismo , DNA Viral/metabolismo , Modelos Animais de Doenças , Regulação Viral da Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/virologia , Vírus Oncolíticos/metabolismo , Regiões Promotoras Genéticas , Simplexvirus/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
6.
Gene Ther ; 11(13): 1023-32, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15164091

RESUMO

The use of viral vectors for gene delivery to motor neurons in vivo has been hampered by the need to perform invasive surgery to inject directly the vector into the anterior horn of the spinal cord. Here, we have characterized the features of herpes simplex virus-1 (HSV)-derived vectors, in terms of gene mutations and promoter constructs, that are required to allow efficient transduction of motor neurons following a relatively noninvasive peripheral administration via sciatic nerve or footpad injection. Owing to the wide variety of animal models used to study neurodegenerative diseases of motor neurons, we analysed the effectiveness of these vectors in adult mice and adult and neonatal rats. We tested viruses with differing degrees of disablement based on the 1764 backbone (deleted for ICP34.5 and an insertional inactivation in VP16) rendered completely replication incompetent by the deletion of the essential immediate-early genes ICP27 and/or ICP4. In the adult mouse, prolonged gene expression in motor neurons was obtained after sciatic nerve inoculation with a vector defective in ICP4 and ICP27. In the adult rat, both the vector defective in ICP4 and the vector defective in ICP4 and ICP27 were capable of transducing motor neurons for extended periods of time during viral latency. This study demonstrates the feasibility of using HSV vectors for persistent transgene expression in motor neurons in a safe and nontoxic manner following peripheral administration. These vectors are potentially useful tools to investigate the functions of genes involved in motor neuronal survival and regeneration in models of motor neuron diseases in vivo.


Assuntos
Gânglios Espinais/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Herpesvirus Humano 1/genética , Neurônios Motores/metabolismo , Doenças Neurodegenerativas/terapia , Animais , Animais Recém-Nascidos , Deleção de Genes , Expressão Gênica , Vetores Genéticos/genética , Humanos , Proteínas Imediatamente Precoces/genética , Injeções , Óperon Lac , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Modelos Animais , Regeneração Nervosa , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático , Fatores de Tempo , Transdução Genética/métodos , Transgenes , Latência Viral
7.
Gene Ther ; 11(10): 829-37, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14985784

RESUMO

Adeno-associated virus (AAV) vectors are highly efficient tools for use in gene therapy. Current production methods rely on plasmid transfection and are not generally considered amenable to scale-up. To improve recombinant AAV (rAAV) vector production in terms of both final titre and simplicity, we constructed recombinant herpes simplex virus (HSV) vectors, either disabled (ICP27 deleted) or nondisabled, encoding the AAV rep and cap genes. We also integrated an rAAVGFP construct into the nondisabled vector and also into a second pair of HSV vectors (disabled and nondisabled) not expressing rep and cap. Transgene incorporation and expression was confirmed by Southern and Western blot, respectively. Optimal double-infection ratios were established for disabled and nondisabled pairs of rep/cap-expressing and rAAVGFP-containing vectors, resulting in up to 1.55 x 10(12) rAAV capsids and 4 x 10(8) expression units from approximately 1 x 10(7) BHK producer cells. Functionality of the prepared vector was confirmed by the detection of abundant green fluorescent protein (GFP) expression following injections of rAAV preparations into the rat brain. This paper therefore describes a simple, efficient, and transfection-free rAAV production process based on the use of HSV and not relying on a proviral cell line that, with appropriate scale-up, could yield quantities of rAAV sufficient for routine clinical use.


Assuntos
Dependovirus/genética , Vetores Genéticos/biossíntese , Simplexvirus/genética , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Recombinação Genética , Simplexvirus/fisiologia , Transfecção , Transgenes , Proteínas Virais/metabolismo , Replicação Viral
8.
Mol Cell Neurosci ; 24(4): 1083-102, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14697671

RESUMO

The expression of Nogo-A mRNA and protein in the nervous system of adult rats and cultured neurons was studied by in situ hybridisation and immunohistochemistry. Nogo-A mRNA was expressed by many cells in unoperated animals, including spinal motor, DRG, and sympathetic neurons, retinal ganglion cells, and neocortical, hippocampal, and Purkinje neurons. Nogo-A protein was strongly expressed by presumptive oligodendrocytes, but not by NG2+glia and was abundant in motor, DRG, and sympathetic neurons, retinal ganglion cells, and many Purkinje cells, but was difficult to detect in dentate gyrus neurons and some neocortical neurons. Cultured fetal mouse neocortical neurons and adult rat DRG neurons strongly expressed Nogo-A in their perikarya, growth cones, and axonal varicosities. All axons in the intact sciatic nerve contained Nogo-A and many but not all regenerating axons were strongly Nogo-A immunopositive after sciatic nerve transection. Ectopic muscle fibres that developed among the regenerating axons were also Nogo-A immunopositive. Following injury to the spinal cord, Nogo-A mRNA was upregulated around the lesion and Nogo-A protein was strongly expressed in injured dorsal column fibres and their sprouts which entered the lesion site. Following optic nerve crush, Nogo-A accumulated in the proximal and distal stumps bordering the lesions.


Assuntos
Proteínas da Mielina/biossíntese , Neurônios/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Neuropatia Ciática/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Proteínas da Mielina/genética , Sistema Nervoso/metabolismo , Proteínas Nogo , Traumatismos do Nervo Óptico/genética , Gravidez , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Neuropatia Ciática/genética , Traumatismos da Medula Espinal/genética
9.
Gene Ther ; 10(4): 292-303, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12595888

RESUMO

Herpes simplex virus type-1 (HSV1) in which the neurovirulence factor ICP34.5 is inactivated has been shown to direct tumour-specific cell lysis in several tumour models. Such viruses have also been shown to be safe in Phase I clinical trials by intra-tumoral injection in glioma and melanoma patients. Previous work has used serially passaged laboratory isolates of HSV1 which we hypothesized may be attenuated in their lytic capability in human tumour cells as compared to more recent clinical isolates. To produce ICP34.5 deleted HSV with enhanced oncolytic potential, we tested two clinical isolates. Both showed improved cell killing in all human tumour cell lines tested compared to a laboratory strain (strain 17+). ICP34.5 was then deleted from one of the clinical isolate strains (strain JS1). Enhanced tumour cell killing with ICP34.5 deleted HSV has also been reported by the deletion of ICP47 by the up-regulation of US11 which occurs following this mutation. Thus to further improve oncolytic properties, ICP47 was removed from JS1/ICP34.5-. As ICP47 also functions to block antigen processing in HSV infected cells, this mutation was also anticipated to improve the immune stimulating properties of the virus. Finally, to provide viruses with maximum oncolytic and immune stimulating properties, the gene for human or mouse GM-CSF was inserted into the JS1/34.5-/47- vector backbone. GM-CSF is a potent immune stimulator promoting the differentiation of progenitor cells into dendritic cells and has shown promise in clinical trials when delivered by a number of means. Combination of GM-CSF with oncolytic therapy may be particularly effective as the necrotic cell death accompanying virus replication should serve to effectively release tumour antigens to then induce a GM-CSF-enhanced immune response. This would, in effect, provide an in situ, patient-specific, anti-tumour vaccine. The viruses constructed were tested in vitro in human tumour cell lines and in vivo in mice demonstrating significant anti-tumour effects. These were greatly improved compared to viruses not containing each of the modifications described. In vivo, both injected and non-injected tumours showed significant shrinkage or clearance and mice were protected against re-challenge with tumour cells. The data presented indicate that JS1/ICP34.5-/ICP47-/GM-CSF acts as a powerful oncolytic agent which may be appropriate for the treatment of a number of solid tumour types in man.


Assuntos
Terapia Genética/métodos , Herpesvirus Humano 1/genética , Imunoterapia/métodos , Neoplasias/terapia , Proteínas Virais/genética , Fatores de Virulência/genética , Animais , Feminino , Deleção de Genes , Engenharia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/imunologia , Transdução Genética
10.
J Virol ; 76(8): 4056-67, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11907244

RESUMO

Herpes simplex virus (HSV) is characterized by its ability to establish a latent infection in sensory neurons, from which it can periodically reactivate. The mechanisms of latency, however, remain unclear. The HSV genome is quiescent during latency except for the expression of the latency-associated transcripts (LATs). Although the exact function of the LATs remains obscure, current evidence suggests they are multifunctional and are involved in both establishment of latency and reactivation from latency. The LATs contain several open reading frames (ORFs). One or more of the functions of the LATs could therefore be protein mediated. We have previously reported that deregulated expression of the largest of the HSV type 1 (HSV-1) LAT ORFs ( approximately 274 amino acids) greatly enhances virus growth in cell types that are normally relatively nonpermissive for HSV replication and also that it complements mutations to the immediate-early (IE) gene ICP0 (S. K. Thomas, G. Gough, D. S. Latchman, and R. S. Coffin, J. Virol. 73:6618-6625, 1999). Here we show that LAT ORF expression overcomes the repression of expression from exogenous promoters introduced into the HSV-1 genome which normally occurs in the absence of IE gene expression. To further explore LAT ORF function, we have generated an epitope-tagged LAT ORF, LATmycHis, which forms punctate structures in the infected-cell nucleus reminiscent of the structures formed by ICP0. These are associated with the appearance of a phosphorylated form of the protein and are formed adjacent to, or around the edges of, viral replication compartments. These results provide further evidence that the HSV-1 LAT ORF protein is biologically functional and that the tightly regulated expression of this protein may be important in the wild-type latency phenotype in vivo.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Genoma Viral , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Fases de Leitura Aberta/genética , Fosforilação , Regiões Promotoras Genéticas , Células Vero , Replicação Viral
11.
J Neurocytol ; 31(2): 93-120, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12815233

RESUMO

At least three proteins present in CNS myelin, Nogo, MAG and OMgp are capable of causing growth cone collapse and inhibiting neurite outgrowth in vitro. Surprisingly, Nogo and OMgp are also strongly expressed by many neurons (including neocortical projection cells). Nogo expression is increased by some cells at the borders of CNS lesion sites and by cells in injured peripheral nerves, but Nogo and CNS myelin are largely absent from spinal cord injury sites, which are none the less strongly inhibitory to axonal regeneration. Nogo is found on growing axons during development, suggesting possible functions for neuronal Nogo in axon guidance. Although Nogo, MAG and OMgp lack sequence homologies, they all bind to the Nogo receptor (NgR), a GPI-linked cell surface molecule which, in turn, binds p75 to activate RhoA. NgR is strongly expressed by cerebral cortical neurons but many other neurons express NgR weakly or not at all. Some neurons, such as DRG cells, respond to Nogo and CNS myelin in vitro although they express little or no NgR in vivo which, with other data, indicates that other receptors are available for NgR ligands. NgR expression is unaffected by injury to the nervous system, and there is no clear correlation between NgR expression by neurons and lack of regenerative ability. In the injured spinal cord, interactions between NgR and its ligands are most likely to be important for limiting regeneration of corticospinal and some other descending tracts; other receptors may be more important for ascending tracts. Antibodies to Nogo, mainly the poorly-characterised IN-1 or its derivatives, have been shown to enhance recovery from partial transections of the spinal cord. They induce considerable plasticity from the axons of corticospinal neurons, including sprouting across the midline and, to a limited extent, regeneration around the lesion. Regeneration of corticospinal axons induced by Nogo antibodies has not yet been demonstrated after complete transections or contusion injuries of the spinal cord. It is not clear whether antibodies against Nogo act on oligodendrocytes/myelin or by binding to neuronal Nogo, or whether they can stimulate regeneration of ascending axons in the spinal cord, most of which express little or no NgR. Despite these uncertainties, however, NgR and its ligands offer important new targets for enhancing plasticity and regeneration in the nervous system.


Assuntos
Axônios/fisiologia , Proteínas da Mielina/biossíntese , Regeneração Nervosa/fisiologia , Medula Espinal/fisiologia , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Ligantes , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Proteínas Nogo
12.
Basic Res Cardiol ; 96(3): 227-36, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11403416

RESUMO

The purpose was to determine the relative efficiency, toxicity and duration of expression following gene delivery by intramyocardial injection of naked DNA, naked DNA complexed to cationic liposomes, naked DNA complexed to cationic liposomes with integrin-targetting peptide, recombinant (E1-/E3-) adenovirus, recombinant adeno-associated virus and recombinant (ICP27-) herpes simplex virus. All vectors incorporated a LacZ reporter driven by a promoter containing the hCMV-IE promoter/enhancer. Efficiency was scored by counting positive cells in five standard microscopic sections harvested from the left ventricular apex. Rabbit hearts (n = 100) were examined from 2 to 56 days after injection. Uncomplexed and complexed naked DNA were very inefficient with less than one positive cell visible per heart. The viral vectors all resulted in robust gene expression with adenovirus being the most efficient by at least one order of magnitude before 21 days. However, despite disparate titres, the efficiency beyond 21 days of adenovirus and adeno-associated virus were comparable. In contrast to adeno-associated virus, both adenovirus and herpes-simplex virus were associated with a marked inflammatory response. Despite reporter gene activity appearing only after 21 days, adeno-associated virus shows comparative promise as a myocardial gene delivery vector.


Assuntos
Técnicas de Transferência de Genes , Miocárdio/metabolismo , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Vírus de DNA/fisiologia , DNA Recombinante/fisiologia , Relação Dose-Resposta a Droga , Regulação Viral da Expressão Gênica/fisiologia , Vetores Genéticos/fisiologia , Inflamação/virologia , Masculino , Modelos Animais , Coelhos , Simplexvirus/genética , Simplexvirus/fisiologia , Transdução Genética
13.
J Virol ; 75(9): 4343-56, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287583

RESUMO

Herpes simplex virus (HSV) has several potential advantages as a vector for delivering genes to the nervous system. The virus naturally infects and remains latent in neurons and has evolved the ability of highly efficient retrograde transport from the site of infection at the periphery to the site of latency in the spinal ganglia. HSV is a large virus, potentially allowing the insertion of multiple or very large transgenes. Furthermore, HSV does not integrate into the host chromosome, removing any potential for insertional activation or inactivation of cellular genes. However, the development of HSV vectors for the central nervous system that exploit these properties has been problematical. This has mainly been due to either vector toxicity or an inability to maintain transgene expression. Here we report the development of highly disabled versions of HSV-1 deleted for ICP27, ICP4, and ICP34.5/open reading frame P and with an inactivating mutation in VP16. These viruses express only minimal levels of any of the immediate-early genes in noncomplementing cells. Transgene expression is maintained for extended periods with promoter systems containing elements from the HSV latency-associated transcript promoter (J. A. Palmer et al., J. Virol. 74:5604-5618, 2000). Unlike less-disabled viruses, these vectors allow highly effective gene delivery both to neurons in culture and to the central nervous system in vivo. Gene delivery in vivo is further enhanced by the retrograde transport capabilities of HSV. Here the vector is efficiently transported from the site of inoculation to connected sites within the nervous system. This is demonstrated by gene delivery to both the striatum and substantia nigra following striatal inoculation; to the spinal cord, spinal ganglia, and brainstem following injection into the spinal cord; and to retinal ganglion neurons following injection into the superior colliculus and thalamus.


Assuntos
Sistema Nervoso Central/virologia , Vírus Defeituosos/fisiologia , Vetores Genéticos/fisiologia , Proteína Vmw65 do Vírus do Herpes Simples/fisiologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Neurônios/virologia , Proteínas Virais/fisiologia , Animais , Transporte Biológico Ativo , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Vírus Defeituosos/genética , Expressão Gênica , Técnicas de Transferência de Genes , Genes Precoces , Genes Reporter , Genes Virais , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Proteína Vmw65 do Vírus do Herpes Simples/genética , Herpesvirus Humano 1/genética , Humanos , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Injeções , Óperon Lac , Proteínas Luminescentes/genética , Masculino , Mutagênese Insercional , Neurônios/citologia , Regiões Promotoras Genéticas , Ratos , Ratos Endogâmicos Lew , Transgenes , Ubiquitina-Proteína Ligases , Células Vero , Proteínas Virais/genética , Proteínas Virais Reguladoras e Acessórias
14.
Gene Ther ; 8(8): 649-53, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11320412

RESUMO

Sustained expression of recombinant proteins is a critical factor for the effectiveness of numerous applications in the biomedical sciences including the treatment of human disease by gene therapy, the large scale production of therapeutic proteins, as well as the investigation of gene function by transgenesis or cell type specific mutagenesis. Although much attention has been paid to the optimisation of regulatory sequences such as promoters, untranslated regions and polyadenylation signals, effective and sustained expression of recombinant genes in vivo is often difficult to achieve. Here we report that the creation of artificial exons, by insertion of two short heterologous introns into open reading frames, is not only compatible with functional expression, but also leads to a 30-fold enhancement of mRNA production for both green fluorescent protein and the bacteriophage P1-derived Cre recombinase. The levels of green fluorescence were increased five-fold in cell lines and sustained long-term expression at increased levels was observed in rat brain after transduction with a herpes simplex virus-based vector. The data presented identify a means by which the expression of recombinant genes can be enhanced considerably, in addition to and independently from the surrounding regulatory sequences. The method should help obtain sustained and effective expression of recombinant proteins in vivo.


Assuntos
Regulação da Expressão Gênica/genética , Terapia Genética/métodos , Íntrons/genética , Animais , Encéfalo/metabolismo , Células CHO , Cricetinae , Éxons/genética , Vetores Genéticos , Proteínas de Fluorescência Verde , Indicadores e Reagentes , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Simplexvirus/genética , Transfecção
15.
Rev Neurosci ; 12(1): 69-78, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11236066

RESUMO

The ability of transplanted neurons from aborted foetuses to produce some therapeutic benefit in Parkinson's disease makes this disease an obvious target for the development of gene therapy procedures which involve delivering the same factors as are provided by the foetal neurons but using a reagent which could be produced in large amounts in a standardised manner. This approach could involve both the delivery of the gene encoding tyrosine hydroxylase to boost dopamine production or the delivery of genes encoding neurotrophic factors such as GDNF to promote the survival of dopaminergic neurons. A variety of different viral and non-viral methods for achieving such gene delivery has been described. These are discussed together with the particular advantages of herpes simplex virus-based vectors which have the potential to deliver multiple therapeutic genes in a single virus vector.


Assuntos
Terapia Genética/métodos , Terapia Genética/tendências , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Doença de Parkinson/genética , Doença de Parkinson/terapia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Humanos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Doença de Parkinson/fisiopatologia , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
16.
Curr Gene Ther ; 1(4): 339-58, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12109061

RESUMO

Herpes simplex virus type 1 (HSV1) has a number of properties which could potentially be exploited in the development of vectors for the delivery of genes to the nervous system. These include a natural tropism for neurons, a large viral genome allowing the insertion of multiple exogenous genes, and the ability to establish asymptomatic life-long latent infections. Despite these inherent advantages, the development of HSV vectors successfully exploiting all these properties has been problematical. This has mainly been due to either vector toxicity or an inability to maintain transgene expression in the long term. Recent progress towards overcoming these problems and several applications of the technology are discussed.


Assuntos
Terapia Genética , Vetores Genéticos , Herpesvirus Humano 1/genética , Neurônios/fisiologia , Animais , Vetores Genéticos/efeitos adversos , Herpesvirus Humano 1/fisiologia , Humanos , Camundongos , Transdução Genética , Latência Viral/genética
17.
Cornea ; 19(3): 369-73, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10832701

RESUMO

PURPOSE: We examined the efficacy and cytopathogenicity of adeno-associated (AAV) and herpes simplex viruses (HSV) as vectors for gene transfer to corneal endothelial cells (CECs). METHODS: Recombinant AAV and HSV were examined for their ability to deliver a lacZ histochemical marker gene to whole-thickness rabbit and human corneas ex vivo. Transgene expression was detected with histochemistry and quantified by a colorimetric assay. RESULTS: Rabbit and human corneas transduced with AAV showed increasing numbers of cells expressing marker gene over a 3- to 4-week period. Using 2.5 x 10(6) or 1.5 x 10(7) infective units for rabbit and human corneal specimens, respectively, approximately 2% of CECs expressed the reporter gene. HSV (10(6) plaque-forming units/specimen) transduced approximately 5% of rabbit and human CECs but showed cytotoxicity. In contrast to the duration of recombinant AAV-mediated lacZ expression, recombinant HSV expression was maximal at day 1 and declined to low levels at day 7. CONCLUSION: AAV is a promising vector, but its usefulness for corneal transduction is currently limited by the technical difficulties preparing high titres. The HSV vector examined is efficient but needs further genetic modification to prolong transgene expression and reduce its toxicity.


Assuntos
Adenoviridae/genética , Endotélio Corneano/enzimologia , Técnicas de Transferência de Genes , Vetores Genéticos , Herpesvirus Humano 1/genética , Óperon Lac/genética , beta-Galactosidase/metabolismo , Animais , Endotélio Corneano/virologia , Expressão Gênica , Histocitoquímica , Humanos , Coelhos
18.
J Virol ; 74(12): 5604-18, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10823868

RESUMO

Herpes simplex virus (HSV) has often been suggested as a suitable vector for gene delivery to the peripheral nervous system as it naturally infects sensory nerve terminals before retrograde transport to the cell body in the spinal ganglia where latency is established. HSV vectors might therefore be particularly appropriate for the study and treatment of chronic pain following vector administration by relatively noninvasive peripheral routes. However parameters allowing safe and efficient gene delivery to spinal ganglia following peripheral vector inoculation, or the long-term expression of delivered genes, have not been comprehensively studied. We have identified combinations of deletions from the HSV genome which allow highly efficient gene delivery to spinal dorsal root ganglia (DRGs) following either footpad or sciatic nerve injection. These vectors have ICP34.5 deleted and have inactivating mutations in vmw65. We also report that peripheral replication is probably necessary for the efficient establishment of latency in vivo, as fully replication-incompetent HSV vectors allow efficient gene expression in DRGs only after peripheral inoculation at a high virus dose. Very low transduction efficiencies are otherwise achieved. In parallel, promoters have been developed that allow the long-term expression of individual or pairs of genes in DRGs by using elements from the latently active region of the virus to confer a long-term activity onto a number of promoters which otherwise function only in the short term. This work further defines elements and mechanisms within the latently active region that are necessary for long-term gene expression and for the first time allows multiple inserted genes to be expressed from HSV vectors during latency.


Assuntos
Técnicas de Transferência de Genes , Engenharia Genética , Vetores Genéticos/genética , Sistema Nervoso Periférico/metabolismo , Simplexvirus/genética , Animais , Linhagem Celular , Cricetinae , Gânglios Espinais/metabolismo , Gânglios Espinais/virologia , Deleção de Genes , Regulação Viral da Expressão Gênica/genética , Genes Reporter/genética , Marcadores Genéticos/genética , Vetores Genéticos/administração & dosagem , Genoma Viral , Proteína Vmw65 do Vírus do Herpes Simples/genética , Membro Posterior/fisiologia , Injeções , Neurônios Motores/metabolismo , Neurônios Motores/virologia , Sistema Nervoso Periférico/virologia , Regiões Promotoras Genéticas/genética , Ribonucleases , Nervo Isquiático/fisiologia , Simplexvirus/patogenicidade , Simplexvirus/fisiologia , Fatores de Tempo , Proteínas Virais/genética , Latência Viral/genética , Replicação Viral/genética
19.
Mov Disord ; 15(1): 9-17, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10634236

RESUMO

Parkinson's disease is an obvious target for the development of gene therapy procedures which could involve both the delivery of the gene encoding tyrosine hydroxylase to boost dopamine production or the delivery of genes encoding neurotrophic factors such as GDNF to promote the survival of dopaminergic neurons. A variety of different viral and nonviral methods for achieving such gene delivery are described together with the particular advantages of herpes simplex virus-based vectors which have the potential to deliver multiple therapeutic genes in a single virus vector.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Fatores de Crescimento Neural , Doença de Parkinson/terapia , Simplexvirus/genética , Animais , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Proteínas do Tecido Nervoso/genética , Doença de Parkinson/genética , Resultado do Tratamento , Tirosina 3-Mono-Oxigenase/genética
20.
J Virol ; 73(9): 7399-409, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438830

RESUMO

Herpes simplex virus (HSV) has often been suggested for development as a vector, particularly for the nervous system. Considerable evidence has shown that for use of HSV as a vector, immediate-early (IE) gene expression must be minimized or abolished, otherwise such vectors are likely to be highly cytotoxic. Mutations of vmw65 which abolish IE promoter transactivating activity may also be included to reduce IE gene expression generally. However, when vmw65 mutations are combined with an IE gene deletion, such viruses are hard to propagate, even on cells which otherwise complement the IE gene deletion effectively. We have found that vmw65 mutants can be effectively grown on cell lines expressing equine herpesvirus 1 gene 12, a non-HSV homologue of vmw65 with little sequence similarity to its HSV counterpart. This prevents repair by homologous recombination of vmw65 mutations in the virus, which would occur if mutations were complemented by vmw65 itself. The gene 12 protein is not packaged into HSV virions, which is important if viruses grown on such cells are to be used as vectors. These results not only further strengthen the evidence for direct functional homology between and similar modes of action of the two proteins but have allowed the generation of gene 12-containing cell lines in which ICP4 and ICP27 expression is induced by virus infection (probably by ICP0) and which give efficient growth of viruses deficient in ICP27, ICP4, and vmw65 (the viruses also have ICP34.5/ORFP deleted). Efficient growth of such viruses has not previously been possible. As these viruses are highly deficient in IE gene expression generally, such virus-cell line combinations may provide an alternative to HSV vectors with deletions of all four of the regulatory IE genes which, for optimal growth, require cell lines containing all four IE genes but which are hard to generate due to the intrinsic cytotoxicity of each of the proteins.


Assuntos
Genes Precoces , Vetores Genéticos , Proteína Vmw65 do Vírus do Herpes Simples/fisiologia , Herpesvirus Equídeo 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Transativadores/fisiologia , Proteínas Virais/fisiologia , Animais , Linhagem Celular , Cricetinae , Deleção de Genes , Teste de Complementação Genética , Vetores Genéticos/genética , Proteína Vmw65 do Vírus do Herpes Simples/genética , Herpesvirus Humano 1/genética , Cavalos , Humanos , Proteínas Imediatamente Precoces/genética , Regiões Promotoras Genéticas , Transativadores/genética , Ativação Transcricional , Ubiquitina-Proteína Ligases , Proteínas Virais/genética , Vírion/fisiologia , Montagem de Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...