Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Bioeng Biotechnol ; 12: 1409203, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38994127

RESUMO

Both cell surface and soluble extracellular glycosaminoglycans have been shown to interfere with the exogenous nucleic acid delivery efficiency of non-viral gene delivery, including lipoplex and polyplex-mediated transfection. Most gene therapy viral vectors used commercially and in clinical trials are currently manufactured using transient transfection-based bioprocesses. The growing demand for viral vector products, coupled with a global shortage in production capability, requires improved transfection technologies and processes to maximise process efficiency and productivity. Soluble extracellular glycosaminoglycans were found to accumulate in the conditioned cell culture medium of suspension adapted HEK293T cell cultures, compromising transfection performance and lentiviral vector production. The enzymatic degradation of specific, chondroitin sulphate-based, glycosaminoglycans with chondroitinase ABC was found to significantly enhance transfection performance. Additionally, we report significant improvements in functional lentiviral vector titre when cultivating cells at higher cell densities than those utilised in a control lentiviral vector bioprocess; an improvement that was further enhanced when cultures were supplemented with chondroitinase ABC prior to transfection. A 71.2% increase in functional lentiviral vector titre was calculated when doubling the cell density prior to transfection compared to the existing process and treatment of the high-density cell cultures with 0.1 U/mL chondroitinase ABC resulted in a further 18.6% increase in titre, presenting a method that can effectively enhance transfection performance.

2.
Biotechnol Bioeng ; 121(9): 2936-2951, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38874319

RESUMO

The demand for Lentiviral Vector (LV) drug substance is increasing. However, primary capture using convective anion-exchange chromatography remains a significant manufacturing challenge. This stems from a poor understanding of the complex adsorption behaviors linked to LVs intricate and variable structure, such as high binding heterogeneity which is typically characterized by a gradient elution profile consisting of two peaks. Understanding which LV structural components drive these phenomena is therefore crucial for rational process design. This work identifies the key LV envelope components responsible for binding to quaternary-amine membrane adsorbents. Eliminating the pseudotype protein (Vesicular Stomatitis Virus G glycoprotein [VSV-G]) did not impact the heterogenous two-peak elution profile, suggesting it is not a major binding species. Digestion of envelope glycosaminoglycans (GAGs), present on proteoglycans, leads to a dramatic reduction in the proportion of vector eluted in peak 2, decreasing from 50% to 3.1%, and a threefold increase in peak 1 maximum. Data from reinjection experiments point towards interparticle envelope heterogeneity from discrete LV populations, where the two-peak profile emerges from a subpopulation of LVs interacting via highly charged GAGs (peak 2) along with a weaker binding population likely interacting through the phospholipid membrane and envelope protein (peak 1).


Assuntos
Vetores Genéticos , Lentivirus , Cromatografia por Troca Iônica/métodos , Lentivirus/genética , Vetores Genéticos/genética , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
3.
Hum Gene Ther ; 29(6): 687-698, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29361840

RESUMO

Due to both the avascularity of the cornea and the relatively immune-privileged status of the eye, corneal transplantation is one of the most successful clinical transplant procedures. However, in high-risk patients, which account for >20% of the 180,000 transplants carried out worldwide each year, the rejection rate is high due to vascularization of the recipient cornea. The main reason for graft failure is irreversible immunological rejection, and it is therefore unsurprising that neovascularization (NV; both pre and post grafting) is a significant risk factor for subsequent graft failure. NV is thus an attractive target to prevent corneal graft rejection. OXB-202 (previously known as EncorStat®) is a donor cornea modified prior to transplant by ex vivo genetic modification with genes encoding secretable forms of the angiostatic human proteins, endostatin and angiostatin. This is achieved using a lentiviral vector derived from the equine infectious anemia virus called pONYK1EiA, which subsequently prevents rejection by suppressing NV. Previously, it has been shown that rabbit donor corneas treated with pONYK1EiA substantially suppress corneal NV, opacity, and subsequent rejection in an aggressive rabbit model of cornea graft rejection. Here, efficacy data are presented in a second rabbit model, which more closely mirrors the clinical setting for high-risk corneal transplant patients, and safety data from a 3-month good laboratory practice toxicology and biodistribution study of pONYK1EiA-modified rabbit corneas in a rabbit corneal transplant model. It is shown that pONYK1EiA-modified rabbit corneas (OXB-202) significantly reduce corneal NV and the rate of corneal rejection in a dose-dependent fashion, and are tolerated with no adverse toxicological findings or significant biodistribution up to 13 weeks post surgery in these rabbit studies. In conclusion, angiogenesis is a valid target to prevent corneal graft rejection in a high-risk setting, and transplanted genetically modified corneas are safe and well-tolerated in an animal model. These data support the evaluation of OXB-202 in a first-in-human trial.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Transplante de Córnea/efeitos adversos , Engenharia Genética , Rejeição de Enxerto/prevenção & controle , Angiostatinas/metabolismo , Animais , Contagem de Células , Neovascularização da Córnea/patologia , Neovascularização da Córnea/terapia , Opacidade da Córnea , Meios de Cultura , Endostatinas/metabolismo , Células Endoteliais/patologia , Feminino , Vetores Genéticos/metabolismo , Rejeição de Enxerto/patologia , Rejeição de Enxerto/fisiopatologia , Células HEK293 , Humanos , Pressão Intraocular , Ceratoplastia Penetrante , Coelhos , Fatores de Risco , Distribuição Tecidual
4.
Mol Ther Methods Clin Dev ; 2: 15017, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26029728

RESUMO

It is a current regulatory requirement to demonstrate absence of detectable replication-competent lentivirus (RCL) in lentiviral vector products prior to use in clinical trials. Immune Design previously described an HIV-1-based integration-deficient lentiviral vector for use in cancer immunotherapy (VP02). VP02 is enveloped with E1001, a modified Sindbis virus glycoprotein which targets dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) expressed on dendritic cells in vivo. Vector enveloped with E1001 does not transduce T-cell lines used in standard HIV-1-based RCL assays, making current RCL testing formats unsuitable for testing VP02. We therefore developed a novel assay to test for RCL in clinical lots of VP02. This assay, which utilizes a murine leukemia positive control virus and a 293F cell line expressing the E1001 receptor DC-SIGN, meets a series of evaluation criteria defined in collaboration with US regulatory authorities and demonstrates the ability of the assay format to amplify and detect a hypothetical RCL derived from VP02 vector components. This assay was qualified and used to test six independent GMP production lots of VP02, in which no RCL was detected. We propose that the evaluation criteria used to rationally design this novel method should be considered when developing an RCL assay for any lentiviral vector.

5.
PLoS One ; 9(4): e94272, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24705452

RESUMO

Usher syndrome type 1B is a combined deaf-blindness condition caused by mutations in the MYO7A gene. Loss of functional myosin VIIa in the retinal pigment epithelia (RPE) and/or photoreceptors leads to blindness. We evaluated the impact of subretinally delivered UshStat, a recombinant EIAV-based lentiviral vector expressing human MYO7A, on photoreceptor function in the shaker1 mouse model for Usher type 1B that lacks a functional Myo7A gene. Subretinal injections of EIAV-CMV-GFP, EIAV-RK-GFP (photoreceptor specific), EIAV-CMV-MYO7A (UshStat) or EIAV-CMV-Null (control) vectors were performed in shaker1 mice. GFP and myosin VIIa expression was evaluated histologically. Photoreceptor function in EIAV-CMV-MYO7A treated eyes was determined by evaluating α-transducin translocation in photoreceptors in response to low light intensity levels, and protection from light induced photoreceptor degeneration was measured. The safety and tolerability of subretinally delivered UshStat was evaluated in macaques. Expression of GFP and myosin VIIa was confirmed in the RPE and photoreceptors in shaker1 mice following subretinal delivery of the EIAV-CMV-GFP/MYO7A vectors. The EIAV-CMV-MYO7A vector protected the shaker1 mouse photoreceptors from acute and chronic intensity light damage, indicated by a significant reduction in photoreceptor cell loss, and restoration of the α-transducin translocation threshold in the photoreceptors. Safety studies in the macaques demonstrated that subretinal delivery of UshStat is safe and well-tolerated. Subretinal delivery of EIAV-CMV-MYO7A (UshStat) rescues photoreceptor phenotypes in the shaker1 mouse. In addition, subretinally delivered UshStat is safe and well-tolerated in macaque safety studies These data support the clinical development of UshStat to treat Usher type 1B syndrome.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Vírus da Anemia Infecciosa Equina/genética , Síndromes de Usher/genética , Síndromes de Usher/terapia , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/metabolismo , Humanos , Macaca , Masculino , Camundongos , Camundongos Knockout , Miosina VIIa , Miosinas/genética , Fenótipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Transporte Proteico , Retina/metabolismo , Retina/patologia , Transducina/metabolismo
6.
Hum Gene Ther ; 25(5): 408-18, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24460027

RESUMO

Corneal transplantation is the oldest and one of the most successful transplant procedures with a success rate in many studies in excess of 90%. The high success rate is mainly attributable to the relatively immune-privileged status of the eye and the fact that the cornea is largely avascular. However, the success rate in patients with failed grafts is much lower such that regrafting is frequently the top indication for corneal transplantation in many centers. Neovascularization is the most important risk factor for rejection, as it allows access of the immune system to the donor tissue, compromising immune privilege of the graft/eye. We have developed a process to modify donor corneal tissue to prevent rejection by a single exposure to a gene therapy vector before surgery (EncorStat(®)). The vector used is based on clinically relevant equine infectious anemia virus (EIAV)-derived lentiviral platform and contains genes for two potently angiostatic genes, endostatin and angiostatin. We show that incubation of rabbit, primate, and human corneal tissue with the EIAV vector mediates strong, stable expression in the corneal endothelium. We have optimized this process to maximize transduction and, once this is complete, maximize the removal of free vector before transplant. Rabbit corneas treated with two different antiangiogenic expression vectors (EIAV-EndoAngio and to a lesser extent EIAV-Endo:k5) significantly suppressed neovascularization in a rabbit model of corneal rejection. As a result, corneal opacity, edema, and inflammatory infiltrates were reduced in these corneas. This study demonstrates that angiogenesis is a suitable target to prevent corneal rejection, and provides the first proof-of-concept data for the development of EncorStat, an ex vivo gene therapy treatment to prevent corneal rejection.


Assuntos
Angiostatinas/uso terapêutico , Neovascularização da Córnea/terapia , Endostatinas/uso terapêutico , Terapia Genética , Vetores Genéticos/metabolismo , Vírus da Anemia Infecciosa Equina/genética , Transdução Genética , Angiostatinas/genética , Animais , Córnea/irrigação sanguínea , Córnea/patologia , Córnea/cirurgia , Neovascularização da Córnea/cirurgia , Opacidade da Córnea , Transplante de Córnea , Endostatinas/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Primatas , Coelhos
7.
Invest Ophthalmol Vis Sci ; 54(6): 4061-71, 2013 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-23620430

RESUMO

PURPOSE: StarGen is an equine infectious anemia virus (EIAV)-based lentiviral vector that expresses the photoreceptor-specific adenosine triphosphate (ATP)-binding cassette transporter (ABCA4) protein that is mutated in Stargardt disease (STGD1), a juvenile macular dystrophy. EIAV vectors are able to efficiently transduce rod and cone photoreceptors in addition to retinal pigment epithelium in the adult macaque and rabbit retina following subretinal delivery. The safety and biodistribution of StarGen following subretinal delivery in macaques and rabbits was assessed. METHODS: Regular ophthalmic examinations, IOP measurements, ERG responses, and histopathology were carried out in both species to compare control and vector-treated eyes. Tissue and fluid samples were obtained to evaluate the persistence, biodistribution, and shedding of the vector following subretinal delivery. RESULTS: Ophthalmic examinations revealed a slightly higher level of inflammation in StarGen compared with control treated eyes in both species. However, inflammation was transient and no overt toxicity was observed in StarGen treated eyes and there were no abnormal clinical findings. There was no StarGen-associated rise in IOP or abnormal ERG response in either rabbits or macaques. Histopathologic examination of the eyes did not reveal any detrimental changes resulting from subretinal administration of StarGen. Although antibodies to StarGen vector components were detected in rabbit but not macaque serum, this immunologic response did not result in any long-term toxicity. Biodistribution analysis demonstrated that the StarGen vector was restricted to the ocular compartment. CONCLUSIONS: In summary, these studies demonstrate StarGen to be well tolerated and localized following subretinal administration.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Vetores Genéticos , Vírus da Anemia Infecciosa Equina/genética , Degeneração Macular/congênito , Células Fotorreceptoras de Vertebrados/metabolismo , Transdução Genética , Animais , Western Blotting , Líquidos Corporais/metabolismo , Citomegalovirus/genética , Eletrorretinografia , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Terapia Genética , Proteínas de Fluorescência Verde/genética , Pressão Intraocular , Macaca mulatta , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/fisiopatologia , Masculino , Reação em Cadeia da Polimerase , Coelhos , Doença de Stargardt , Distribuição Tecidual , Transfecção
8.
Hum Gene Ther Methods ; 23(5): 309-23, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23121195

RESUMO

The release of lentiviral vectors for clinical use requires the testing of vector material, production cells, and, if applicable, ex vivo-transduced cells for the presence of replication-competent lentivirus (RCL). Vectors derived from the nonprimate lentivirus equine infectious anemia virus (EIAV) have been directly administered to patients in several clinical trials, with no toxicity observed to date. Because EIAV does not replicate in human cells, and because putative RCLs derived from vector components within human vector production cells would most likely be human cell-tropic, we previously developed an RCL assay using amphotropic murine leukemia virus (MLV) as a surrogate positive control and human cells as RCL amplification/indicator cells. Here we report an additional RCL assay that tests for the presence of theoretical "equine-tropic" RCLs. This approach provides further assurance of safety by detecting putative RCLs with an equine cell-specific tropism that might not be efficiently amplified by the human cell-based RCL assay. We tested the ability of accessory gene-deficient EIAV mutant viruses to replicate in a highly permissive equine cell line to direct our choice of a suitable EIAV-derived positive control. In addition, we report for the first time the mathematical rationale for use of the Poisson distribution to calculate minimal infectious dose of positive control virus and for use in monitoring assay positive/spike control failures in accumulating data sets. No RCLs have been detected in Good Manufacturing Practice (GMP)-compliant RCL assays to date, further demonstrating that RCL formation is highly unlikely in contemporary minimal lentiviral vector systems.


Assuntos
Vetores Genéticos/genética , Vírus da Anemia Infecciosa Equina/genética , Tropismo Viral , Replicação Viral , Animais , Bioensaio , Linhagem Celular , Ordem dos Genes , Cavalos , Humanos , Vírus da Anemia Infecciosa Equina/fisiologia , Vírus da Leucemia Murina , Camundongos , Reprodutibilidade dos Testes , Transdução Genética
9.
Development ; 135(13): 2289-99, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18508860

RESUMO

The outgrowth of the vertebrate tail is thought to involve the proliferation of regionalised stem/progenitor cell populations formed during gastrulation. To follow these populations over extended periods, we used cells from GFP-positive transgenic chick embryos as a source for donor tissue in grafting experiments. We determined that resident progenitor cell populations are localised in the chicken tail bud. One population, which is located in the chordoneural hinge (CNH), contributes descendants to the paraxial mesoderm, notochord and neural tube, and is serially transplantable between embryos. A second population of mesodermal progenitor cells is located in a separate dorsoposterior region of the tail bud, and a corresponding population is present in the mouse tail bud. Using heterotopic transplantations, we show that the fate of CNH cells depends on their environment within the tail bud. Furthermore, we show that the anteroposterior identity of tail bud progenitor cells can be reset by heterochronic transplantation to the node region of gastrula-stage chicken embryos.


Assuntos
Neurônios/metabolismo , Células-Tronco/metabolismo , Cauda/embriologia , Cauda/metabolismo , Animais , Animais Geneticamente Modificados , Contagem de Células , Diferenciação Celular , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Neurônios/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Cauda/citologia
10.
J Gene Med ; 9(5): 345-56, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17366519

RESUMO

Pseudotyping viral vectors with vesicular stomatitis virus glycoprotein (VSV-G) enables the transduction of an extensive range of cell types from different species. We have discovered two important parameters of the VSV-G-pseudotyping phenomenon that relate directly to the transduction potential of lentiviral vectors: (1) the glycosylation status of VSV-G, and (2) the quantity of glycoprotein associated with virions. We measured production-cell and virion-associated quantities of two isoform variants of VSV-G, which differ in their glycosylation status, VSV-G1 and VSV-G2, and assessed the impact of this difference on the efficiency of mammalian cell transduction by lentiviral vectors. The glycosylation of VSV-G at N336 allowed greater maximal expression of VSV-G in HEK293T cells, thus facilitating vector pseudotyping. The transduction of primate cell lines was substantially affected (up to 50-fold) by the degree of VSV-G1 or VSV-G2 incorporation, whereas other cell lines, such as D17 (canine), were less sensitive to virion-associated VSV-G1/2 quantities. These data indicate that the minimum required concentration of virion-associated VSV-G differs substantially between cell species/types. The implications of these data with regard to VSV-G-pseudotyped vector production, titration, and use in host-cell restriction studies, are discussed.


Assuntos
Vetores Genéticos , Lentivirus/genética , Glicoproteínas de Membrana/genética , Transdução Genética , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular , Glicosilação , Humanos , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/química , Isoformas de Proteínas/análise , Isoformas de Proteínas/química , Especificidade da Espécie , Proteínas do Envelope Viral/análise , Proteínas do Envelope Viral/química , Proteínas Virais/análise , Proteínas Virais/química , Proteínas Virais/genética , Vírion/química
11.
Mol Ther ; 12(3): 537-46, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16099415

RESUMO

The use of lentiviral vectors for gene transfer into hematopoietic stem cells has raised considerable interest as these vectors can permanently integrate their genome into quiescent cells. Vectors based on alternative lentiviruses would theoretically be safer than HIV-1-based vectors and could also be used in HIV-positive patients, minimizing the risk of generating replication-competent virus. Here we report the use of third-generation equine infectious anemia virus (EIAV)- and HIV-1-based vectors with minimal viral sequences and absence of accessory proteins. We have compared their efficiency in transducing mouse and human hematopoietic stem cells both in vitro and in vivo to that of a previously documented second-generation HIV-1 vector. The third-generation EIAV- and HIV-based vectors gave comparable levels of transduction and transgene expression in both mouse and human NOD/SCID repopulating cells but were less efficient than the second-generation HIV-1 vector in human HSCs. For the EIAV vector this is possibly a reflection of the lower protein expression levels achieved in human cells, as vector copy number analysis revealed that this vector exhibited a trend to integrate equally efficiently compared to the third-generation HIV-1 vector in both mouse and human HSCs. Interestingly, the presence or absence of Tat in viral preparations did not influence the transduction efficiency of HIV-1 vectors in human HSCs.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/instrumentação , Vetores Genéticos , HIV/genética , Células-Tronco Hematopoéticas/metabolismo , Vírus da Anemia Infecciosa Equina/genética , Animais , Antígenos CD34/biossíntese , Linhagem Celular , Citometria de Fluxo , Terapia Genética/métodos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Técnicas In Vitro , Lentivirus/genética , Lentivirus/metabolismo , Antígenos Comuns de Leucócito/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Células-Tronco/citologia
12.
Blood ; 104(9): 2714-21, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15231566

RESUMO

Hemophilia B, also known as Christmas disease, arises from mutations in the factor IX (F9) gene. Its treatment in humans, by recombinant protein substitution, is expensive, thus limiting its application to intermittent treatment in bleeding episodes and prophylaxis during surgery; development of inhibitory antibodies is an associated hazard. This study demonstrates permanent therapeutic correction of his disease without development of immune reactions by introduction of an HIV-based lentiviral vector encoding the human factor IX protein into the fetal circulation of immunocompetent hemophiliac and normal outbred mice. Plasma factor IX antigen remained at around 9%, 13%, and 16% of normal in the 3 hemophilia B mice, respectively, until the last measurement at 14 months. Substantial improvement in blood coagulability as measured by coagulation assay was seen in all 3 mice and they rapidly stopped bleeding after venipuncture. No humoral or cellular immunity against the protein, elevation of serum liver enzymes, or vector spread to the germline or maternal circulation were detected.


Assuntos
Fator IX/administração & dosagem , Terapias Fetais/métodos , Terapia Genética/métodos , Hemofilia B/terapia , Animais , Coagulação Sanguínea/efeitos dos fármacos , Fator IX/genética , Fator IX/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Tolerância Imunológica , Imunocompetência , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos , Fenótipo , Circulação Placentária , Gravidez
13.
Nature ; 429(6990): 413-7, 2004 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15164063

RESUMO

Amyotrophic lateral sclerosis (ALS) causes adult-onset, progressive motor neuron degeneration in the brain and spinal cord, resulting in paralysis and death three to five years after onset in most patients. ALS is still incurable, in part because its complex aetiology remains insufficiently understood. Recent reports have indicated that reduced levels of vascular endothelial growth factor (VEGF), which is essential in angiogenesis and has also been implicated in neuroprotection, predispose mice and humans to ALS. However, the therapeutic potential of VEGF for the treatment of ALS has not previously been assessed. Here we report that a single injection of a VEGF-expressing lentiviral vector into various muscles delayed onset and slowed progression of ALS in mice engineered to overexpress the gene coding for the mutated G93A form of the superoxide dismutase-1 (SOD1(G93A)) (refs 7-10), even when treatment was only initiated at the onset of paralysis. VEGF treatment increased the life expectancy of ALS mice by 30 per cent without causing toxic side effects, thereby achieving one of the most effective therapies reported in the field so far.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Transporte Axonal , Modelos Animais de Doenças , Vírus da Anemia Infecciosa Equina/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Esclerose Lateral Amiotrófica/patologia , Animais , Tronco Encefálico/patologia , Progressão da Doença , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Injeções Intramusculares , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação Puntual/genética , Medula Espinal/patologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Taxa de Sobrevida , Fatores de Tempo , Transgenes/genética , Fator A de Crescimento do Endotélio Vascular/efeitos adversos , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Mol Ther ; 9(1): 101-11, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14741783

RESUMO

We have developed a non-primate-based lentiviral vector based on the equine infectious anemia virus (EIAV) for efficient gene transfer to the central and peripheral nervous systems. Previously we have demonstrated that pseudotyping lentiviral vectors with the rabies virus glycoprotein confers retrograde axonal transport to these vectors. In the present study we have successfully produced high-titer EIAV vectors pseudotyped with envelope glycoproteins from Rhabdovirus vesicular stomatitis virus (VSV) serotypes (Indiana and Chandipura strains); rabies virus [various Evelyn-Rokitnicki-Abelseth ERA strains and challenge virus standard (CVS)]; Lyssavirus Mokola virus, a rabies-related virus; and Arenavirus lymphocytic choriomeningitis virus (LCMV). These vectors were delivered to the striatum or spinal cord of adult rats or muscle of neonatal mice by direct injection. We report that the lentiviral vectors pseudotyped with envelopes from the VSV Indiana strain, wild-type ERA, and CVS strains resulted in strong transduction in the striatum, while Mokola- and LCMV-pseudotyped vectors exhibited moderate and weak transduction, respectively. Furthermore ERA- and CVS-pseudotyped lentiviral vectors demonstrated retrograde transport and expression in distal neurons after injection in brain, spinal cord, and muscle. The differences in transduction efficiencies and retrograde transport conferred by these envelope glycoproteins present novel opportunities in designing therapeutic strategies for different neurological diseases.


Assuntos
Vetores Genéticos , Vírus da Anemia Infecciosa Equina/genética , Sistema Nervoso/metabolismo , Transdução Genética , Proteínas do Envelope Viral/genética , Animais , Transporte Biológico , Corpo Estriado/citologia , Expressão Gênica/fisiologia , Terapia Genética , Glicoproteínas/metabolismo , Injeções , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Lyssavirus/genética , Masculino , Camundongos , Músculo Esquelético , Doenças do Sistema Nervoso/terapia , Vírus da Raiva/genética , Vírus da Raiva/metabolismo , Ratos , Ratos Wistar , Medula Espinal , Proteínas do Envelope Viral/metabolismo
15.
Virology ; 314(2): 488-96, 2003 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-14554078

RESUMO

Expression of the murine leukaemia virus (MLV) major Gag antigen p65(Gag) using the baculovirus expression system leads to efficient assembly and release of virus-like particles (VLP) representative of immature MLV. Expression of p180(Gag-Pol), facilitated normally in mammalian cells by readthrough of the p65(Gag) termination codon, also occurs efficiently in insect cells to provide a source of the MLV protease and a pattern of p65(Gag) processing similar to that observed in mammalian cells. VLP release from p180(Gag-Pol)-expressing cells however remains essentially immature with disproportionate levels of the uncleaved p65(Gag) precursor when compared to the intracellular Gag profile. Changing the p65(Gag) termination codon altered the level of p65(Gag) and p180(Gag-Pol) within expressing cells but did not alter the pattern of released VLP, which remained immature. Coexpression of p65(Gag) with a fixed readthrough p180(Gag-Pol) also led to only immature VLP release despite high intracellular protease levels. Our data suggest a mechanism that preferentially selects uncleaved p65(Gag) for the assembly of MLV in this heterologous expression system and implies that, in addition to their relative levels, active sorting of the correct p65(Gag) and p180(Gag-Pol) ratios may occur in producer cells.


Assuntos
Vírus da Leucemia Murina/metabolismo , Vírion/metabolismo , Montagem de Vírus , Animais , Células Cultivadas , Proteínas de Fusão gag-pol/metabolismo , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina/genética , Microscopia Eletrônica , Nucleopoliedrovírus/genética , Nucleopoliedrovírus/metabolismo , Spodoptera/virologia , Vírion/ultraestrutura
16.
Nat Biotechnol ; 21(5): 569-72, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12679787

RESUMO

Human immunodeficiency virus type 1 (HIV-1)-based vectors are currently made by transient transfection, or using packaging cell lines in which expression of HIV-1 Gag and Pol proteins is induced. Continuous vector production by cells in which HIV-1 Gag-Pol is stably expressed would allow rapid and reproducible generation of large vector batches. However, attempts to make stable HIV-1 packaging cells by transfection of plasmids encoding HIV-1 Gag-Pol have resulted in cells which secrete only low levels of p24 antigen (20-80 ng/ml), possibly because of the cytotoxicity of HIV-1 protease. Infection of cells with HIV-1 can result in stable virus production; cell clones that produce up to 1,000 ng/ml secreted p24 antigen have been described. Here we report that expression of HIV-1 Gag-Pol by a murine leukemia virus (MLV) vector allows constitutive, long-term, high-level (up to 850 ng/ml p24) expression of HIV-1 Gag. Stable packaging cells were constructed using codon-optimized HIV-1 Gag-Pol and envelope proteins of gammaretroviruses; these producer cells could make up to 10(7) 293T infectious units (i.u.)/ml (20 293T i.u./cell/day) for at least three months in culture.


Assuntos
Regulação Viral da Expressão Gênica/genética , Vetores Genéticos/biossíntese , Vetores Genéticos/genética , HIV-1/crescimento & desenvolvimento , HIV-1/genética , Transfecção/métodos , Vetores Genéticos/isolamento & purificação , HIV-1/isolamento & purificação , Células HeLa , Humanos , Controle de Qualidade
17.
Mol Ther ; 5(5 Pt 1): 566-70, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11991747

RESUMO

Ideally, gene transfer vectors used in clinical protocols should only express the gene of interest. So far most vectors have contained marker genes to aid their titration. We have used quantitative real-time PCR to titrate equine infectious anemia virus (EIAV) vectors for gene therapy applications. Viral RNA was isolated from vector preparations and analyzed in a one-step RT-PCR reaction in which reverse transcription and amplification were combined in one tube. The PCR assay of vector stocks was quantitative and linear over four orders of magnitude. In tandem, the integration efficiency of these vectors has also been determined by real-time PCR, measuring the number of vector genomes in the target cells. We have found that these methods permit reliable and sensitive titration of lentiviral vectors independent from the expression of a transgene. They also allow us to determine the integration efficiency of different vector genomes. This technology has proved very useful, especially in the absence of marker genes and where vectors express multiple genes.


Assuntos
Vetores Genéticos , Vírus da Anemia Infecciosa Equina/genética , Animais , Linhagem Celular , Citomegalovirus/genética , Primers do DNA/química , DNA Viral/metabolismo , Expressão Gênica , Terapia Genética , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , RNA Viral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA