Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Cancer Gene Ther ; 19(12): 888-98, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23099884

RESUMO

Despite the tremendous potential of adenovirus (Ad) as a delivery vector for cancer gene therapy, its use in clinical settings has been limited, mainly as a result of the limited infectivity in many tumors and the wide tissue tropism associated with Ad. To modify the tropism of the virus, we have inserted the epidermal growth factor-like domain of the human heregulin-α (HRG) into the HI loop of Ad5 fiber. This insertion had no adverse effect on fiber trimerization nor did it affect incorporation of the modified fiber into infectious viral particles. Virions bearing modified fiber displayed growth characteristics and viral yields indistinguishable from those of wild-type (wt) virus. Most importantly, HRG-tagged virions showed enhanced infection of cells expressing the cognate receptors HER3/ErbB3 and HER4/ErbB4. This was significantly reduced in the presence of soluble HRG. Furthermore, HER3-expressing Chinese hamster ovary (CHO) cells were transduced by the HRG-modified virus, but not by wt virus. In contrast, CHO cells expressing the coxsackie-Ad receptor were transduced with both viruses. However, infection of an in vivo breast cancer xenograft model after intratumoral injection was similar with both viruses, suggesting that the tumor microenvironment and/or the route of delivery have important roles in infection of target cells with fiber-modified Ads.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/genética , Neoplasias da Mama/virologia , Receptor ErbB-3/metabolismo , Adenoviridae/metabolismo , Animais , Neoplasias da Mama/metabolismo , Cricetinae , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Injeções Intralesionais , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Gene Ther ; 11(6): 504-11, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14999222

RESUMO

Helper-dependent adenovirus vectors (hdAd), which are deleted of all viral protein-coding sequences, can mediate long-term expression of a therapeutic transgene and lead to life-long, phenotypic correction in animal models of genetic disease. Here, we describe a new system for the generation of hdAd, which utilizes the DNA size restrictions imposed on an Ad virion deleted of protein IX (pIX): such virions are reported to package up to only approximately 35 kb of viral DNA. A pIX(-) helper virus (approximately 37.3 kb) was easily grown on complementing 293pIX cells. Upon infection of noncomplementing cells, this virus was not capable of forming infectious virions, but provided replicative and packaging functions for propagation of a 30-kb hdAd. The pIX(-) helper virus was effective in amplifying an hdAd and, in combination with Cre-mediated excision in the viral-packaging signal, resulted in a 1000-fold reduction in helper virus contamination in hdAd stocks compared to Cre/lox alone, as determined by plaque assay. However, through slot blot analysis of DNA isolated from virions, we determined that the ratio of hdAd to helper DNA was 500:1, similar to the ratio observed when using Cre/lox alone. Surprisingly, a large amount of the 37.3-kb helper DNA was being packaged into the pIX-deleted virions, but these virions were incapable of establishing productive infections in plaque assays, for reasons which are still unclear. Nevertheless, the pIX(-) hdAd generated in this system infected cells and expressed a transgene at levels similar to those obtained with a pIX(+) hdAd. These data suggest that, although further studies are necessary to characterize the nature of the defective helper virions formed in this system, deletion of pIX from the helper virus genome does provide an effective method to prevent recovery of functional helper virus during hdAd amplification.


Assuntos
Adenoviridae/genética , Proteínas do Capsídeo/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Vírus Auxiliares/genética , Carcinoma/terapia , Linhagem Celular Tumoral , Deleção de Genes , Genes Virais , Humanos , Neoplasias Pulmonares/terapia , Inativação de Vírus , Replicação Viral
3.
J Thromb Haemost ; 2(1): 111-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14717974

RESUMO

BACKGROUND: Adenoviral-based methods of gene therapy have been ineffective at providing sustained factor (F)VIII expression in outbred populations of large animal hemophilic models primarily due to the immunogenicity of these vectors. Improvements have been made in vector design leading to the development of the helper-dependent adenoviral (HD) system. Unfortunately, it remains unclear whether these modifications are sufficient to circumvent the induction of inhibitor formation associated with adenoviral gene transfer. OBJECTIVE: To develop an HD vector capable of mediating sustained FVIII expression and to determine the variables that influence inhibitor development. METHODS: HD vectors were constructed encoding the canine FVIII B-domain deleted transgene under the control of either the cytomegalovirus (CMV) promoter or a tissue-restricted hybrid element consisting of five HNF-1 binding sites, located upstream of the human FVIII proximal promoter. Inbred and outbred populations of hemophilic mice were treated, and monitored for vector-induced toxicity, therapeutic efficacy, and inhibitor formation. RESULTS: When HD vectors utilizing the CMV promoter were administered, all hemophilic mice developed high levels of FVIII inhibitors. In contrast, vectors under the control of the HNF/FVIII element were capable of achieving sustained elevations of FVIII for over 6 months. Strain-specific differences were also observed, with outbred animals showing a greater propensity towards inhibitor development in response to treatment. CONCLUSIONS: HD vectors can be used to provide long-term FVIII expression in hemophilic animals, but treatment outcome and the induction of inhibitors is dependent on a number of variables including the transgene promoter, the vector dose, and the genetic background of the host.


Assuntos
Terapia Genética/métodos , Hemofilia A/terapia , Adenoviridae/genética , Animais , Sequência de Bases , DNA Recombinante/genética , Modelos Animais de Doenças , Cães , Fator VIII/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos , Vírus Auxiliares/genética , Hemofilia A/genética , Hemofilia A/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/genética , Distribuição Tecidual
4.
Cancer Gene Ther ; 8(5): 361-70, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11477456

RESUMO

Synergy between interleukin-12 (IL-12) and B7-1 (CD80) for cancer immunotherapy has previously been demonstrated in animal models of breast cancer, lymphoma, and multiple myeloma. With a view to human clinical application, tricistronic retroviral and adenovirus vectors co-expressing IL-12 (IL-12p40 plus IL-12p35) and CD80 were constructed by utilizing two internal ribosome entry site (IRES) sequences to link the three cDNAs. A murine stem cell virus (MSCV)-based retroviral vector (MSCV-hIL12.B7) utilized distinct IRES sequences from the encephalomyocarditis virus (EMCV) and the foot-and-mouth disease virus (FMCV), whereas Ad5-based adenovirus vectors contained transcriptional units with two EMCV IRES sequences under the control of murine (AdMh12.B7) or human (AdHh12.B7) cytomegalovirus promoters. AdMh12.B7 was found to consistently direct higher levels of IL-12 and CD80 expression than AdHh12.B7 following infection of a number of human tumor cell lines. In preclinical studies, the human myeloma cell line U266 was infected with MSCV-hIL12.B7 and a resulting clonal cell line, U/MSCV-h12.B7, was generated with stable expression of CD80 and secreting IL-12 at 1 ng/24 h/10(6) cells. By comparison, following AdMh12.B7 infection, 81% of infected U266 cells (U/AdMh12.B7) expressed CD80 and secreted IL-12 at 25-50 ng/24 h/10(6) cells. Both engineered myeloma cell lines stimulated enhanced allogeneic mixed lymphocyte proliferation and provoked increases in cytotoxic T-lymphocyte responses and gamma-interferon release from normal donor lymphocytes exposed to parental U266 cells. These results suggest potential clinical utility of AdMh12.B7 in immunotherapy strategies for the treatment of multiple myeloma and other cancers.


Assuntos
Adenoviridae/genética , Antígeno B7-1/genética , Vetores Genéticos , Imunoterapia/métodos , Interleucina-12/genética , Neoplasias/terapia , Retroviridae/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Antígeno B7-1/metabolismo , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica , DNA Complementar , Quimioterapia Combinada , Citometria de Fluxo , Técnicas de Transferência de Genes , Humanos , Interleucina-12/metabolismo , Neoplasias/metabolismo , Neoplasias/virologia , Linfócitos T/imunologia , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/virologia
5.
Mol Ther ; 3(5 Pt 1): 809-15, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11356086

RESUMO

Helper-dependent (HD) adenoviral vectors devoid of all viral coding sequences have a large cloning capacity and have been reported to provide long-term transgene expression in vivo with negligible toxicity, making them attractive vectors for gene therapy. Currently, the most efficient means of generating HD vectors involves co-infecting 293 cells expressing Cre with the HD vector and a helper virus bearing a packaging signal flanked by loxP sites. Cre-mediated excision of the packaging signal renders the helper virus genome unpackageable but still able to replicate and to provide helper functions for HD vector propagation. HD vector titer is increased by serial co-infections. Typically, helper virus contamination is < or =1% pre- and < or =0.1% postpurification by CsCl banding. While these contamination levels are low, further reduction is desirable. Alternative methods of selection against the helper virus may achieve this goal, especially when combined with Cre/loxP. We describe the development of a system for generating HD vectors based on site-specific recombination between frt sites catalyzed by FLP recombinase and show by direct comparison that the FLP/frt and Cre/loxP systems are equivalent with respect to HD vector amplification efficiency and helper virus contamination levels. Availability of a second recombinase system for HD vector production will enhance the utility and flexibility of HD vectors.


Assuntos
Adenoviridae/genética , DNA Nucleotidiltransferases/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Vírus Auxiliares/genética , Proteínas Virais , Linhagem Celular , DNA/metabolismo , Humanos , Integrases/genética , Modelos Genéticos , Plasmídeos/metabolismo , Transgenes
6.
Leukemia ; 15(5): 846-54, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11368448

RESUMO

Eight multiple myeloma patients participated in a phase I trial evaluating the feasibility and safety of subcutaneous vaccination with adenovirus engineered, autologous plasma cells after high-dose therapy. Plasma cells were concentrated from bone marrow harvests by negative selection and high gradient magnetic separation. The mean plasma cell yield was 2.61 x 10(8). Transgene expression measured 48 h after plasma cell infection with an IL-2 expressing adenovirus averaged 2.95 ng/ml/10(6) cells. Vaccine production was successful for 88% of patients. Two months after high-dose therapy, six patients received from one to five injections of 3.5-9.0 x 10(7) cells/vaccine. Vaccines were well tolerated with only minor systemic symptoms reported. Injection with tumor cells induced a local inflammatory response consisting predominantly of CD8+ and/or TIA-1+ T-lymphocytes. Myeloma specific anti-tumor responses, assessed by interferon-gamma (IFN-gamma) release and cytotoxic T cell killing of autologous tumor cells, were not enhanced after vaccination in one evaluable patient. Clinical response, manifested as a decrease in serum paraprotein, was not observed in the one patient who had measurable disease at the time of vaccination. These results demonstrate that the generation of adenovector modified plasma cell vaccines is technically feasible and can be safely administered post-transplant. Further studies of immunlogic and clinical efficacy are required.


Assuntos
Terapia Genética , Interleucina-2/genética , Mieloma Múltiplo/terapia , Plasmócitos/imunologia , Vacinação , Adenoviridae/genética , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
7.
J Natl Cancer Inst ; 93(6): 472-9, 2001 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-11259473

RESUMO

BACKGROUND: Cytokine-based gene therapy strategies efficiently stimulate immune responses against many established transplanted tumors, leading to rejection of the tumor. In this study, we investigated the therapeutic potential of cancer immunotherapy in a clinically more relevant model, woodchucks with primary hepatocellular carcinomas induced by woodchuck hepatitis virus. METHODS: Large (2-5 cm), established intrahepatic tumors were given an injection once with 1 x 10(9) plaque-forming units of AdIL-12/B7.1, an adenovirus vector carrying genes for murine interleukin 12 and B7.1, or of AdEGFP, the control virus, and regression of the tumors was then monitored. Five animals were used in total. RESULTS: In four tumor-bearing animals, the antitumor response was assessed by autopsy and histologic analysis within 1-2 weeks after treatment. In all animals treated with AdIL-12/B7.1 therapy versus AdEGFP therapy, we observed substantial tumor regression (P =.006; two-sided unpaired Student's t test) accompanied by a massive infiltration of T lymphocytes. These tumors also contained increased levels of CD4(+) and CD8(+) T cells and interferon gamma (IFN gamma). In continuously growing tumor nodules given an injection of the control virus or in nontumoral liver, no such effects (i.e., tumor regression and increased levels of CD4(+) and CD8(+) T cells and IFN gamma) were detected. In the fifth animal, monitored for long-term antitumor efficacy by magnetic resonance imaging (MRI) after intratumoral vector administration by MRI guidance, the tumor was almost completely eliminated (> or = 95%) 7 weeks after treatment. CONCLUSION: Adenovirus vector-based immunotherapy appears to be an effective treatment of large nontransplanted (orthotopic) tumors that acquire malignant characteristics in a stepwise process, reflecting the real-world scenario of hepatocellular carcinoma in humans.


Assuntos
Adenoviridae , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Imunoterapia/métodos , Interleucina-12/administração & dosagem , Interleucina-12/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Vetores Genéticos , Hepatite Viral Animal/complicações , Interferon gama/análise , Interleucina-12/genética , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Marmota
8.
Immunology ; 101(3): 388-96, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11106943

RESUMO

In this study, we have examined the in vivo effects of interleukin-5 (IL-5) and IL-6 over-expression on systemic and mucosal immune responses using recombinant human type 5 adenoviruses capable of expressing these cytokines upon infection. A recombinant adenovirus containing the murine IL-5 gene within the E3 region was constructed and found to express high levels of IL-5 protein both in vitro and in vivo. Intranasal inoculation of mice with this vector or a vector expressing murine IL-6 increased adenovirus-specific immunoglobulin A (IgA) titres in lung lavage fluid threefold compared with those elicited by control virus. The simultaneous expression of both cytokines by co-inoculation altered the kinetics of the mucosal anti-adenovirus IgA response and resulted in a more than additive increase in antibody titres. The co-expression effect on IgA synthesis was not due to an increase in numbers of antigen-specific resident lung tissue lymphocytes. When mucosal IgG responses were examined, IL-6 expression had the largest impact on anti-adenovirus levels, whereas co-expression produced an intermediate response. Systemic immune responses were also affected by IL-6 expression as a twofold increase in serum IgG anti-adenovirus titres was observed after a secondary challenge with wild-type adenovirus. These results demonstrate a relevant role for IL-5 and IL-6 in the development of mucosal immune responses in vivo and suggest that the incorporation of either IL-5 and/or IL-6 into recombinant adenovirus vectors may be a useful tool in the development of mucosal vaccines.


Assuntos
Adenovírus Humanos/imunologia , Imunoglobulina A Secretora/biossíntese , Interleucinas/imunologia , Pulmão/imunologia , Vacinas Virais/imunologia , Administração Intranasal , Animais , Células Produtoras de Anticorpos/imunologia , Feminino , Vetores Genéticos/imunologia , Imunidade nas Mucosas , Imunização/métodos , Interleucina-5/imunologia , Interleucina-6/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL
10.
Biotechniques ; 29(3): 524-6, 528, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10997266

RESUMO

We have recently developed a high-efficiency method of constructing adenovirus vectors based on Cre-mediated recombination between two plasmids co-transfected into 293 cells. The simplicity and efficiency of this method should greatly expedite the construction of most recombinant vectors. However, this system would not be suitable for constructing vectors bearing loxP sites elsewhere in the genome because of undesirable Cre-mediated vector rearrangements. To address this, we have developed a similar system using FLP-mediated site-specific recombination for the construction of adenovirus vectors.


Assuntos
Adenoviridae/genética , DNA Nucleotidiltransferases/metabolismo , Vetores Genéticos , Saccharomyces cerevisiae/enzimologia , Proteínas Virais , Linhagem Celular , DNA/genética , Humanos , Integrases/metabolismo , Plasmídeos/genética , Transfecção , beta-Galactosidase/genética
11.
Mol Med ; 6(3): 179-95, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10965494

RESUMO

BACKGROUND: Certain gene therapy protocols may require multiple administrations of vectors to achieve therapeutic benefit to the patient. This may be especially relevant for vectors such as adenoviral vectors that do not integrate into the host chromosome. Because immunocompetent animal models used for gene transfer studies develop neutralizing antibodies to adenoviral vectors after a single administration, little is known about how repeat administrations of vectors might affect transgene expression and vector toxicity. MATERIALS AND METHODS: We used mice deficient in the membrane spanning region of immunoglobulin (IgM), which do not develop antibodies, to evaluate the effect of repeated intravenous administration of first-generation and helper-dependent adenoviral vectors expressing human alpha 1-antitrypsin (hAAT). The duration and levels of transgene expression were evaluated after repeated administration of vectors. Toxicity was assessed by measuring the level of liver enzymes in the serum and the degrees of hepatocyte hypertrophy and proliferation. RESULTS: We found that previous administration of first-generation adenoviral vectors can alter the response to subsequent doses. These alterations included an increase in transgene expression early (within 1 and 3 days), followed by a rapid drop in expression by day 7. In addition, previous administrations of first-generation vectors led to an increase in toxicity of subsequent doses, as indicated by a rise in liver enzymes and an increase in hepatocyte proliferation. In contrast to first-generation vectors, use of the helper-dependent adenovirus vector, Ad-STK109, which contained no viral coding regions, did not lead to increased toxicity after multiple administrations. CONCLUSIONS: We conclude that the response of the host to adenoviral vectors can be altered after repeated administration, compared with the response after the initial vector dose. In addition, these experiments provide further evidence for the relative safety of helper-dependent adenoviral vectors for gene therapy, compared with first-generation vectors.


Assuntos
Adenoviridae/genética , Terapia Genética/efeitos adversos , Vetores Genéticos , Vírus Auxiliares/genética , Animais , Expressão Gênica , Terapia Genética/métodos , Proteínas de Homeodomínio/genética , Humanos , Imunoglobulina M/genética , Fígado/enzimologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Segurança , alfa 1-Antitripsina/genética
13.
Gene Ther ; 7(14): 1200-9, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10918488

RESUMO

Recombinant adenovirus vectors have provided a major advance in gene delivery systems for post-mitotic neurons. However, the use of these first generation vectors has been limited due to the onset of virally mediated effects on cellular function and viability. In the present study we have used primary cultures of cerebellar granule neurons to examine the efficacy and cytotoxic effects of a helper-dependent adenovirus vector (hdAd) in comparison with a first generation vector. Our results demonstrate that the hdAd system provides equally efficient infectivity with significantly reduced toxicity in comparison to first generation vectors. Neurons transduced with a high titre of a first generation vector exhibited a time-dependent shut down in global protein synthesis and impaired physiological function as demonstrated by a loss of glutamate receptor responsiveness. This was followed by an increase in the fraction of TUNEL-positive cells and a loss of neuronal survival. In contrast, hdAds could be used at titres that transduce >85% of neurons with little cytotoxic effect: cellular glutamate receptor responses and rates of protein synthesis were indistinguishable from uninfected controls. Furthermore, cell viability was not significantly affected for at least 7 days after infection. At excessive viral titres, however, infection with hdAd did cause moderate but significant changes in cell function and viability in primary neuronal cultures. Thus, while these vectors are remarkably improved over first generation vectors, these also have limitations with respect to viral effects on cellular function and viability. Gene Therapy (2000) 7, 1200-1209.


Assuntos
Adenoviridae/genética , Vetores Genéticos/uso terapêutico , Neurônios , Animais , Sobrevivência Celular , Células Cultivadas , Vetores Genéticos/efeitos adversos , Camundongos , N-Metilaspartato/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Biossíntese de Proteínas , Transdução Genética/genética
14.
Hum Mol Genet ; 9(7): 1109-17, 2000 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-10767336

RESUMO

Adult onset primary open angle glaucoma is a leading cause of blindness throughout the world. The disease results in an apoptotic death of retinal ganglion cells that is usually associated with an elevation of intraocular pressure. Familial aggregation of the disorder provides evidence for strong genetic influences that are likely to be the result of multiple susceptibility genes. A two-stage genome scan to identify the genomic locations of glaucoma susceptibility genes was performed using an initial pedigree set of 113 affected sibpairs and a second pedigree set of 69 affected sibpairs. Linkage analysis was performed using both model-dependent (lod score) and model-independent affected relative pair and sibpair methods. Twenty-five regions identified by the initial scan were further investigated using the second pedigree set. In the combined data analysis, regions located on chromosomes 2, 6, 9, 11, 14, 17 and 19 continued to produce model-dependent lod scores and/or an MLS >1.0, while five regions (2, 14, 17p, 17q and 19) produced an MLS >2. 0. Multipoint analysis using ASPEX also showed significant results on chromosomes 2, 14, 17p, 17q and 19. These results are an important step towards the identification of genes responsible for the genetic susceptibility to this blinding condition.


Assuntos
Mapeamento Cromossômico , Glaucoma de Ângulo Aberto/genética , Idade de Início , Cromossomos , Saúde da Família , Genes Dominantes , Genes Recessivos , Predisposição Genética para Doença , Genoma Humano , Genótipo , Humanos , Escore Lod , Repetições de Microssatélites
15.
Hum Gene Ther ; 11(5): 693-9, 2000 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10757349

RESUMO

The two-plasmid rescue method of constructing Ad vectors, which relies on either homologous or Cre-mediated recombination between two plasmids cotransfected into 293 or 293Cre4 cells, respectively, offers advantages over other approaches because of its simplicity. We have improved the efficiency of vector construction by both homologous and Cre-mediated recombination by replacing the single ITR in the shuttle plasmid with a head-to-head ITR junction. We have also expanded the versatility of this method by incorporating a Cre expression cassette into the plasmids to permit high-efficiency Cre-mediated vector rescue using 293 cells, abrogating the need for Cre-expressing cell lines. This new system retains the simplicity of the original but results in an approximately 100-fold increase in the number of recombinant viruses produced, all of which contain the foreign DNA insert, and allows high-efficiency Cre-mediated vector isolation using any E1-complementing cell line.


Assuntos
Adenoviridae/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Plasmídeos/genética , Proteínas Virais , Linhagem Celular/virologia , Humanos , Integrases/genética , Recombinação Genética , Sequências Repetidas Terminais
16.
J Dent Res ; 79(2): 701-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10728970

RESUMO

Adenoviral vectors effectively transfer genes to rat salivary glands. However, potent immune responses limit their use in vivo. Mice offer more opportunities than rats for the study of these immune processes. We first established conditions for infection of mouse salivary glands, with an adenoviral vector. The effects of time, viral dose, viral diluent buffer volume, and dexamethasone on expression of a transgene, luciferase, were determined by means of the recombinant vector AdCMVluc. Optimal luciferase expression was observed when the vector was suspended in 50 microL of buffer. This volume completely filled the gland parenchyma and slightly distended the capsule. Dexamethasone increased immediate transgene expression and reduced the acute inflammation one day following viral administration, but did not alter subsequent mononuclear inflammation or transgene expression 14 or 28 days later. An adenoviral vector encoding either anti-inflammatory cytokine IL-4 or IL-10 was co-administered with AdCMVluc to increase transgene expression at 14 and 28 days. While this strategy did not extend the duration of luciferase expression, co-administration of AdCMVIL-10 with AdCMVluc almost completely eliminated the chronic inflammatory infiltrate in the glands after 28 days. This study demonstrates that adenoviral-mediated gene transfer to mouse submandibular glands is possible by intraductal cannulation and that reduction of either the acute or chronic inflammatory infiltrates was insufficient to increase long-term transgene expression in this tissue.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Glândula Submandibular/metabolismo , Adjuvantes Imunológicos/genética , Animais , Anti-Inflamatórios/uso terapêutico , Soluções Tampão , Dexametasona/uso terapêutico , Feminino , Seguimentos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Glucocorticoides/uso terapêutico , Interleucina-10/genética , Interleucina-4/genética , Luciferases/genética , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Sialadenite/genética , Sialadenite/prevenção & controle , Glândula Submandibular/enzimologia , Glândula Submandibular/imunologia , Doenças da Glândula Submandibular/genética , Doenças da Glândula Submandibular/prevenção & controle , Fatores de Tempo
17.
Hum Gene Ther ; 10(16): 2667-72, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10566894

RESUMO

Adenovirus (Ad) vectors provide a highly efficient means of mammalian gene transfer and are widely used for high-level protein expression in mammalian cells, as recombinant vaccines and for gene therapy. A commonly used method for constructing Ad vectors relies on in vivo homologous recombination between two Ad DNA-containing bacterial plasmids cotransfected into 293 cells. While the utility of this two-plasmid approach is well established, its efficiency is low owing to the inefficiency of homologous recombination. To address this, we have developed an improved method for Ad vector construction based on Cre-mediated site-specific recombination between two bacterial plasmids, each bearing a loxP site. Ad vectors are generated as a result of Cre-mediated site-specific recombination between the two plasmids after their cotransfection into 293 cells expressing Cre recombinase. The frequency of Ad vector rescue by Cre-mediated site-specific recombination is significantly higher (approximately 30-fold) than by in vivo homologous recombination. The efficiency and reliability of this method should greatly simplify and expedite the construction of recombinant Ad vectors for mammalian gene transfer. Ad vectors are commonly constructed by homologous recombination between two plasmids cotransfected into 293 cells. This method has numerous advantages but results in low numbers of plaques owing to inefficient recombination. We have developed an improved method based on Cre-mediated site-specific recombination, which results in vector rescue at frequencies approximately 30-fold higher than by homologous recombination. This method should greatly simplify and expedite the construction of recombinant Ad vectors for mammalian gene transfer.


Assuntos
Adenoviridae/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Integrases/genética , Proteínas Virais , Sequência de Bases , Linhagem Celular , Técnicas de Transferência de Genes , Vetores Genéticos/biossíntese , Humanos , Integrases/metabolismo , Dados de Sequência Molecular , Plasmídeos/genética , Recombinação Genética , Transfecção
18.
Cancer Gene Ther ; 6(5): 465-74, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10505857

RESUMO

The toxic effects of tumor necrosis factor alpha (TNFalpha) have greatly limited its use in tumor therapy. Recently, clear evidence has been obtained linking the p55 TNF receptor (TNFR) to the induction of systemic toxicity. We have generated a p75 murine TNFR (mTNFR)-specific mutant of mTNFalpha (D142N-A144R), cloned this gene into a recombinant adenovirus vector (Ad-75), and studied its efficacy for tumor immunotherapy of a murine transgenic breast cancer model. Cell culture supernatants from Ad-75-transduced cells showed no cytotoxic activity on L929 cells, but retained the ability to induce proliferation of a murine T-cell line (CT6); this activity was not blocked by soluble p55 mTNFR. Furthermore, it was shown that the mutant form of mTNFalpha was able to coimmunoprecipitate only with the p75 mTNFR and not with the p55 mTNFR. Tumors injected with Ad-75 became necrotic, and mice injected with < or =1 x 10(9) plaque-forming units showed no mortality, whereas both wild-type murine and human TNF vectors induced lethality at doses of 1 and 5 x 10(8) plaque-forming units. All Ad-TNF vectors induced partial or permanent tumor regressions, with cured mice showing immune memory against the tumor. These results demonstrate that a p75 mTNFR agonist expressed from a recombinant adenovirus vector does not induce mortality at doses that cause tumor regression.


Assuntos
Adenoviridae/genética , Antígenos CD/genética , Vetores Genéticos , Neoplasias Mamárias Experimentais/imunologia , Receptores do Fator de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/genética , Animais , Sequência de Bases , Divisão Celular , Primers do DNA , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Mutação , Receptores Tipo II do Fator de Necrose Tumoral
19.
J Immunol ; 163(8): 4546-56, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10510398

RESUMO

Rheumatoid arthritis is a chronic inflammatory joint disease, leading to cartilage and bone destruction. In this study, we investigated the effects of local IL-4 application, introduced by a recombinant human type 5 adenovirus vector, in the knee joint of mice with collagen-induced arthritis. One intraarticular injection with an IL-4-expressing virus caused overexpression of IL-4 in the mouse knee joint. Enhanced onset and aggravation of the synovial inflammation were found in the IL-4 group. However, despite ongoing inflammation, histologic analysis showed impressive prevention of chondrocyte death and cartilage erosion. In line with this, chondrocyte proteoglycan synthesis was enhanced in the articular cartilage. This was quantified with ex vivo 35S-sulfate incorporation in patellar cartilage and confirmed by autoradiography on whole knee joint sections. Reduction of cartilage erosion was further substantiated by lack of expression of the stromelysin-dependent cartilage proteoglycan breakdown neoepitope VDIPEN in the Ad5E1 mIL-4-treated knee joint. Reduced metalloproteinase activity was also supported by markedly diminished mRNA expression of stromelysin-3 in the synovial tissue. Histologic analysis revealed marked reduction of polymorphonuclear cells in the synovial joint space in the IL-4-treated joints. This was confirmed by immunolocalization studies on knee joint sections using NIMP-R14 staining and diminished mRNA expression of macrophage-inflammatory protein-2 in the synovium tissue. mRNA levels of TNF-alpha and IL-1beta were suppressed as well, and IL-1beta and nitric oxide production by arthritic synovial tissue were strongly reduced. Our data show an impressive cartilage-protective effect of local IL-4 and underline the feasibility of local gene therapy with this cytokine in arthritis.


Assuntos
Adenoviridae/genética , Artrite Experimental/prevenção & controle , Cartilagem Articular/patologia , Colágeno/imunologia , Vetores Genéticos/imunologia , Membro Posterior/imunologia , Interleucina-4/biossíntese , Articulações/imunologia , Adenoviridae/imunologia , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Artrite Experimental/patologia , Cartilagem Articular/imunologia , Cartilagem Articular/metabolismo , Morte Celular/imunologia , Movimento Celular/imunologia , Condrócitos/imunologia , Condrócitos/metabolismo , Condrócitos/patologia , Regulação para Baixo/imunologia , Epitopos/biossíntese , Granulócitos/imunologia , Granulócitos/patologia , Membro Posterior/metabolismo , Membro Posterior/patologia , Injeções Intra-Articulares , Interleucina-1/antagonistas & inibidores , Interleucina-1/biossíntese , Interleucina-4/administração & dosagem , Articulações/metabolismo , Articulações/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Oligopeptídeos/biossíntese , Fragmentos de Peptídeos/biossíntese , Membrana Sinovial/imunologia , Membrana Sinovial/patologia
20.
Proc Natl Acad Sci U S A ; 96(22): 12816-21, 1999 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-10536005

RESUMO

The efficiency of first-generation adenoviral vectors as gene delivery tools is often limited by the short duration of transgene expression, which can be related to immune responses and to toxic effects of viral proteins. In addition, readministration is usually ineffective unless the animals are immunocompromised or a different adenovirus serotype is used. Recently, adenoviral vectors devoid of all viral coding sequences (helper-dependent or gutless vectors) have been developed to avoid expression of viral proteins. In mice, liver-directed gene transfer with AdSTK109, a helper-dependent adenoviral (Ad) vector containing the human alpha(1)-antitrypsin (hAAT) gene, resulted in sustained expression for longer than 10 months with negligible toxicity to the liver. In the present report, we have examined the duration of expression of AdSTK109 in the liver of baboons and compared it to first-generation vectors expressing hAAT. Transgene expression was limited to approximately 3-5 months with the first-generation vectors. In contrast, administration of AdSTK109 resulted in transgene expression for longer than a year in two of three baboons. We have also investigated the feasibility of circumventing the humoral response to the virus by sequential administration of vectors of different serotypes. We found that the ineffectiveness of readministration due to the humoral response to an Ad5 first-generation vector was overcome by use of an Ad2-based vector expressing hAAT. These data suggest that long-term expression of transgenes should be possible by combining the reduced immunogenicity and toxicity of helper-dependent vectors with sequential delivery of vectors of different serotypes.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Fígado/metabolismo , Animais , Divisão Celular , Vetores Genéticos/imunologia , Vírus Auxiliares/genética , Humanos , Masculino , Camundongos , Testes de Neutralização , Papio , Baço/citologia , Baço/metabolismo , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...