Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Protoc Hum Genet ; Chapter 12: Unit 12.3, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18428249

RESUMO

This unit describes preparation of adenovirus-polylysine-DNA complexes, which is useful for transfection of DNA into a variety of cell types. A DNA complex is prepared with biotinylated adenovirus and streptavidin-polylysine, coupled to transferrin, and used to transfect cells. Several support protocols describe methods for adenovirus growth and purification, biotinylation, inactivation with psoralen, and quantitation of the adenovirus particles. Additional support protocols describes preparation of streptavidin-polylysine and transferrin-polylysine, necessary for the basic procedure. The DNA used for transfection must be free of lipopolysaccharide (LPS), and two methods for removing LPS are described. A more direct polylysine-virus linkage that is simple and requires no exotic reagents can be used for transfection. This protocol requires polylysine-modified adenovirus, prepared as described. An alternate protocol describes transfecting cells with free virus and DNA condensed with a polycation.


Assuntos
Adenoviridae/genética , Adenoviridae/isolamento & purificação , DNA Recombinante/genética , DNA Recombinante/isolamento & purificação , Polilisina/isolamento & purificação , Animais , Linhagem Celular , Técnicas Genéticas , Genética Médica , Humanos , Substâncias Macromoleculares/isolamento & purificação , Transfecção
2.
Hum Gene Ther ; 10(6): 983-93, 1999 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-10223732

RESUMO

We performed a phase I trial to evaluate the safety and tolerability of repeated skin injections of IL-2-transfected autologous melanoma cells into patients with advanced disease. Cell suspensions, propagated from excised metastases, were IL-2 gene transfected by adenovirus-enhanced transferrinfection and X-irradiated prior to injection. Vaccine production was successful in 54% of the patients. Fifteen patients (37%) received two to eight skin vaccinations of either 3 x 10(6) (intradermal) or 1 x 10(7) (half intradermal, half subcutaneous) transfected melanoma cells per vaccination (secreting 140-17,060 biological response modifier program units of IL-2/10(6) cells/24 hr). Analyses of safety and efficacy were carried out in 15 and 14 patients, respectively. Overall, the vaccine was well tolerated. All patients displayed modest local reactions (erythema, induration, and pruritus) and some experienced flu-like symptoms. Apart from newly appearing (4 of 14) and increasing (5 of 14) anti-adenovirus and newly detectable anti-nuclear antibody titers (1 of 15), recipients developed de novo or exhibited increased melanoma cell-specific delayed-type hypersensitivity (DTH) reactions (8 of 15) and vitiligo (3 of 15) and showed signs of tumor regression (3 of 15). This supports the idea of a vaccine-induced or -amplified anti-cancer immune response. None of the patients exhibited complete or partial regressions, but five of them experienced periods of disease stabilization. Three of these individuals received more than the four planned vaccinations and their mean survival time was 15.7 +/- 3.5 months as compared to 7.8 +/- 4.6 months for the entire patient cohort. These data indicate that IL-2-producing, autologous cancer cells can be safely administered to stage IV melanoma patients and could conceivably be of benefit to patients with less advanced disease.


Assuntos
Vacinas Anticâncer/uso terapêutico , Melanoma/terapia , Neoplasias Cutâneas/terapia , Adulto , Idoso , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Feminino , Humanos , Hipersensibilidade Tardia , Injeções Intralesionais , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias Cutâneas/patologia , Resultado do Tratamento
4.
Gene ; 190(1): 211-6, 1997 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-9185869

RESUMO

Cancer vaccines are based on the concept that tumors express novel antigens and thus differ from their normal tissue counterparts. Such putative tumor-specific antigens should be recognizable by the immune system. However, malignant cells are of self origin and only poorly immunogenic, which limits their capability to induce an anticancer immune response. To overcome this problem, tumor cells have been isolated, genetically engineered to secrete cytokine gene products and administered as cancer vaccines. We used adenovirus-enhanced transferrinfection (AVET), which allows high-level transient transgene expression, to introduce cytokine gene expression vectors into murine melanoma cells. The efficiency of AVET makes laborious selection and cloning procedures obsolete. We administered such modified tumor cells as cancer vaccines to syngeneic animals and investigated their impact on the induction of anticancer immunity. We found that IL-2 or GM-CSF gene-transfected murine melanoma cells are highly effective vaccines. Both of these cytokine-secreting vaccines cured 80% of animals which bore a subcutaneous micrometastasis prior to treatment, and induced potent antitumor immunity. The generation of antitumor immunity by these cytokine-secreting vaccines requires three different steps: (1) tumor antigen uptake and processing by antigen-presenting cells (APCs) at the site of vaccination; (2) migration of these APCs into the regional lymph nodes where T-cell priming occurs; (3) recirculation of specific, activated T-cells that recognize distinct tumor load and initiate its elimination. Extending our previously reported studies, we have now comprehensively analysed the requirements for effective antitumor vaccination in animals. This may also become the basis for treatment of human cancer patients.


Assuntos
Vacinas Anticâncer/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-2/metabolismo , Transfecção , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Interleucina-2/genética , Linfonodos/imunologia , Melanoma Experimental/terapia , Camundongos , Neoplasia Residual/terapia
5.
Proc Natl Acad Sci U S A ; 94(7): 3256-61, 1997 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9096380

RESUMO

The discovery of a steadily growing number of tumor antigens (TAs) has made generic, cell-free, peptide-based cancer vaccines a possible alternative to cytokine-transfected autologous cellular cancer vaccines. The major drawback of peptide vaccines, however, is the poor immunogenicity of peptides. It is commonly thought that for the induction of an effective anticancer immune response, antigen-presenting cells (APCs) have to display TA-derived peptides to T lymphocytes. Polycationic amino acids have been employed in the past to enhance transport of proteins into cells. In a systematic study, the ability of different cationic polymers to transfer fluorescence-tagged peptides to APCs was investigated. We were able to show that several compounds enhance uptake of fluorescence-labeled peptides by APCs to different degrees. The most efficient compound identified, polyarginine (pArg), enhanced peptide delivery by more than 2 logs as compared with cells treated with peptide alone, whereas polylysine (pLys) treatment resulted in approximately 10-fold increased levels of fluorescence. Augmentation of peptide uptake was concentration-dependent, and the molecular weight of pArg or pLys also influenced peptide delivery. Furthermore, highly negatively charged peptides appear to be delivered with higher efficiency, although neutral peptides were also taken up at enhanced rates. Whereas peptide uptake mediated by pLys appears to be due to an at least transient permeabilization of cell membranes, peptide delivery in the presence of pArg may rely on endocytic processes. TA-derived peptides applied as cancer vaccines in conjunction with polycations afforded antitumor protection in animal models.


Assuntos
Células Apresentadoras de Antígenos/química , Antígenos de Neoplasias/química , Peptídeos/administração & dosagem , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/enzimologia , Cátions , Linhagem Celular , L-Lactato Desidrogenase/metabolismo , Camundongos , Dados de Sequência Molecular
6.
Proc Natl Acad Sci U S A ; 94(7): 3262-7, 1997 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9096381

RESUMO

The central role that tumor antigen-derived peptides play in induction of antitumor immunity makes them ideal candidates for peptide-based cancer vaccines. We have demonstrated that "transloading" is an efficient strategy for importing short peptide ligands into antigen-presenting cells in vitro. Postulating that the transloading procedure might effect peptide uptake by antigen-presenting cells in vivo as well, we tested this approach for the generation of peptide-based cancer vaccines. In the P815 mastocytoma system, we vaccinated mice by s.c. injection of a single, known natural peptide derived from JAK-1 kinase. Whereas vaccination with peptide alone or mixed with incomplete Freund's adjuvant was ineffective, application of the peptide in conjunction with the polycation poly-L-lysine protected a significant number of animals against tumor challenge. Dependent upon the type of poly-L-lysine applied, protection against tumor take was comparable to that achieved with irradiated whole-cell vaccines, genetically modified to secrete granulocyte-macrophage colony-stimulating factor. In the murine melanoma M-3, a combination of four putative tumor antigen-derived peptides was tested as a cancer vaccine. Administered in combination with polycations, these peptides evoked potent antitumor immunity that could not be obtained with the peptides alone or peptides emulsified in incomplete Freund's adjuvant. However, peptide-polycation vaccines applied to the M-3 model were not as efficient as cellular control vaccines, consisting of irradiated interleukin 2 or granulocyte-macrophage colony-stimulating factor-secreting tumor cells.


Assuntos
Antígenos de Neoplasias/química , Vacinas Anticâncer/química , Peptídeos/química , Sequência de Aminoácidos , Animais , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Sistema Livre de Células , Sarcoma de Mastócitos/prevenção & controle , Melanoma Experimental/prevenção & controle , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos DBA , Dados de Sequência Molecular
8.
Mol Cell Biol ; 17(3): 1189-200, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9032246

RESUMO

The cleavage-stage (CS) histones of the sea urchin are known to be maternally expressed in the egg, have been implicated in chromatin remodeling of the male pronucleus following fertilization, and are the only histone variants present in embryonic chromatin up to the four-cell stage. With the help of partial peptide sequence information, we have isolated and identified CS H1, H2A, H2B, H3, and H4 cDNAs from egg poly(A)+ mRNA of the sea urchin Psammechinus miliaris. All five CS proteins correspond to replacement histone variants which are encoded by replication-independent genes containing introns, poly(A) addition signals, and long nontranslated sequences. Transcripts of the CS histone genes could be detected only during oogenesis and in development up to the early blastula stage. The CS proteins, with the exception of H4, are unique histones which are distantly related in sequence to the early, late, and sperm histone subtypes of the sea urchin. In contrast, the CS H1 protein displays highest sequence homology with the H1M (B4) histone of Xenopus laevis. Both H1 proteins are replacement histone variants with very similar developmental expression profiles in their respective species, thus indicating that the frog H1M (B4) gene is a vertebrate homolog of the CS H1 gene. These data furthermore suggest that the CS histones are of ancient evolutionary origin and may perform similar conserved functions during oogenesis and early development in different species.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Ouriços-do-Mar/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar/genética , Proteínas do Ovo/genética , Embrião não Mamífero/química , Feminino , Genes/genética , Variação Genética , Histonas/análise , Masculino , Dados de Sequência Molecular , Família Multigênica/genética , Oogênese , RNA Mensageiro/genética , Ouriços-do-Mar/embriologia , Homologia de Sequência de Aminoácidos , Xenopus laevis/embriologia
9.
Gene Ther ; 3(9): 819-24, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8875231

RESUMO

It has been postulated that IL-2 secreting cancer vaccines establish antitumor immunity because the cytokine acting in a paracrine fashion would deliver a helper signal directly to the T cells making contact with the modified tumor cells at the site of vaccination. However, patterns of lymphocyte recirculation cannot be reconciled with the above direct interaction model: only primed memory T cells rather than naive T lymphocytes patrol the periphery, while naive T cells travel to the peripheral lymph nodes, where priming occurs. We have found that in vivo treatment of mice with the antibody MEL-14 directed against L-selectin, which is a molecule expressed at high levels on naive T cells, can completely abrogate protection against a mouse melanoma conferred by an IL-2 secreting vaccine. Since mouse memory CD4 and CD8 T cells are L-selectin-low, only migration of naive T cells is perturbed by the in vivo antibody blockade. Thus, priming of naive T cells in the draining lymph node is a critical step for the successful vaccination by IL-2 secreting cancer vaccines. Such priming is performed most efficiently by professional antigen presenting cells; consequently, these data also imply that allogeneic origin of tumor vaccines may not exclude successful vaccination.


Assuntos
Vacinas Anticâncer/imunologia , Interleucina-2/metabolismo , Linfonodos/imunologia , Melanoma Experimental/prevenção & controle , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais , Células Apresentadoras de Antígenos/imunologia , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/genética , Memória Imunológica , Interleucina-2/genética , Selectina L/imunologia , Lectinas Tipo C , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos DBA , RNA Mensageiro/análise , Receptores de Interleucina-2/genética , Células Tumorais Cultivadas , Vacinação
10.
Proc Natl Acad Sci U S A ; 93(18): 9759-63, 1996 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-8790404

RESUMO

The major hurdle to be cleared in active immunotherapy of cancer is the poor immunogenicity of cancer cells. In previous attempts to overcome this problem, whole tumor cells have been used as vaccines, either admixed with adjuvant(s) or genetically engineered to express nonself proteins or immunomodulatory factors before application. We have developed a novel approach to generate an immunogeneic, highly effective vaccine: major histocompatibility complex (MHC) class I-positive cancer cells are administered together with MHC class I-matched peptide ligands of foreign, nonself origin, generated by a procedure we term transloading. Murine tumor lines of the H2-Kd or the H2-Db haplotype, melanoma M-3 and B16-F10, respectively, as well as colon carcinoma CT-26 (H2-Kd), were transloaded with MHC-matched influenza virus-derived peptides and applied as irradiated vaccines. Mice bearing a deposit of live M-3 melanoma cells were efficiently cured by this treatment. In the CT-26 colon carcinoma and the B16-F10 melanoma, high efficacies were obtained against tumor challenge, suggesting the universal applicability of this new type of vaccine. With foreign peptide ligands adapted to the requirements of a desired MHC class I haplotype, this concept may be used for the treatment of human cancers.


Assuntos
Antígenos H-2/imunologia , Neoplasias/prevenção & controle , Vacinas/biossíntese , Animais , Antígenos Virais/imunologia , Linhagem Celular , Neoplasias do Colo/imunologia , Neoplasias do Colo/prevenção & controle , Citometria de Fluxo , Humanos , Ligantes , Melanoma/imunologia , Melanoma/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias/imunologia , Orthomyxoviridae
11.
Cytokines Mol Ther ; 2(3): 185-91, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9384703

RESUMO

Cancer vaccines are genetically modified tumor cells that, by cytokine secretion or by expression of costimulatory molecules, are capable of mobilizing the host's immune system to destroy tumor cells. We have used adenovirus-enhanced transferrinfection (AVET) for the generation of cancer vaccines. This is a highly efficient method to deliver various genes into a large proportion of tumor cells, making further selection unnecessary. We found in the mouse M-3 melanoma model that two consecutive vaccinations with transfected cells secreting IL-2 protect animals from tumor development by a subsequent challenge, and result in long-lasting tumor-specific immunity dependent on both CD4+ and CD8+ T cells. Patterns of lymphocyte recirculation and the need for CD4+ T cells indicated that the role of IL-2 is not merely local 'replacement of help', as has been proposed before. Instead, our findings suggest a three-stage process for the generation of effector T cells after vaccination with IL-2 secreting tumor cells: (1) tumor antigen uptake and processing at the site of injection by APCs, (2) migration of APCs into the regional draining lymph nodes where T-cell priming occurs, and (3) recirculation of activated cytotoxic T cells, that recognize and eliminate distant tumor cells. This model also implies that allogeneic tumor cells or synthetic tumor antigens may be used with success in future cancer vaccines.


Assuntos
Vacinas Anticâncer/biossíntese , Melanoma Experimental/terapia , Transfecção/métodos , Adenoviridae , Animais , Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Terapia Genética/métodos , Vetores Genéticos , Humanos , Interleucina-2/biossíntese , Melanoma Experimental/imunologia , Camundongos , Polilisina , Linfócitos T Citotóxicos/imunologia , Transferrina/biossíntese
13.
Hum Gene Ther ; 6(6): 753-61, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7548275

RESUMO

Retrovirus-mediated gene transfer is currently the method of choice for the transfection of human T lymphocytes for applications in gene therapy. Use of retroviral vectors, however, is hampered by limits on the size of the genetic material to be transferred, the requirement of dividing target cells, and by potential safety questions. Synthetic peptide-enhanced or adenovirus-enhanced receptor-mediated transferrinfection of DNA (SPET and AVET, respectively) is a powerful method for the introduction of genetic material into mammalian cells. Although transferrin has proven to be a useful ligand for gene transfer in many cell types, gene expression in T cells with transferrin/DNA complexes is usually not satisfactory. To improve gene transfer to T cells, antibodies directed against the CD3-T cell receptor complex were tested for their ability to function as ligands for DNA delivery. In T cell lines, up to 50% of the cells expressed a beta-galactosidase reporter gene using anti-CD3 gene transfer complexes. Applying optimized conditions, prestimulated primary peripheral blood lymphocytes were also transfected successfully, although at a lesser efficiency (5%).


Assuntos
Anticorpos/metabolismo , DNA Recombinante/metabolismo , Técnicas de Transferência de Genes , Complexo Receptor-CD3 de Antígeno de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Anticorpos/genética , Anticorpos/imunologia , Endocitose , Humanos , Complexo Receptor-CD3 de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Células Tumorais Cultivadas , beta-Galactosidase/genética
14.
Proc Natl Acad Sci U S A ; 92(12): 5540-4, 1995 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-7777545

RESUMO

Although both CD4+ and CD8+ T cells are clearly required to generate long-lasting anti-tumor immunity induced by s.c. vaccination with interleukin 2 (IL-2)-transfected, irradiated M-3 clone murine melanoma cells, some controversy continues about the site and mode of T-cell activation in this system. Macrophages, granulocytes, and natural killer cells infiltrate the vaccination site early after injection into either syngeneic euthymic DBA/2 mice or athymic nude mice and eliminate the inoculum within 48 hr. We could not find T cells at the vaccination site, which argues against the concept that T-cell priming by the IL-2-secreting cancer cells occurs directly at that location. However, reverse transcription-PCR revealed transcripts indicative of T-cell activation and expansion in the draining lymph nodes of mice immunized with the IL-2-secreting vaccine but not in mice vaccinated with untransfected, irradiated M-3 cells. We therefore propose that the antigen-presenting cells, which invade the vaccination site, process tumor-derived antigens and, subsequently, initiate priming of tumor-specific T lymphocytes in lymphoid organs. These findings suggest a three-stage process for the generation of effector T cells after vaccination with IL-2-secreting tumor cells: (i) tumor-antigen uptake and processing at the site of injection by antigen-presenting cells, (ii) migration of antigen-presenting cells into the regional draining lymph nodes, where T-cell priming occurs, and (iii) circulation of activated T cells that either perform or initiate effector mechanisms leading to tumor cell destruction.


Assuntos
Interleucina-2/metabolismo , Linfonodos/patologia , Melanoma Experimental/imunologia , Linfócitos T/imunologia , Vacinas/imunologia , Animais , Sequência de Bases , Citocinas/genética , Citocinas/metabolismo , Primers do DNA , Interleucina-2/uso terapêutico , Linfonodos/metabolismo , Ativação Linfocitária , Macrófagos/ultraestrutura , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Camundongos Nus , Microscopia Eletrônica , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas , Vacinação
15.
Proc Natl Acad Sci U S A ; 92(10): 4711-4, 1995 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-7753870

RESUMO

Cancer vaccines genetically engineered to produce interleukin 2 have been investigated intensively in a series of animal models and are at the point of entering into clinical trials. In this study we demonstrate a strong correlation between the rate of interleukin 2 production and the protection efficiency of murine S91 melanoma cell (clone M-3) vaccines. Best immunization is achieved with vaccines producing medium interleukin 2 levels of 1000-3000 units per 10(5) cells per day. Reduced interleukin 2 production evokes a corresponding decline in the number of successfully treated animals. Unexpectedly, when interleukin 2 expression is raised to high levels of 5000-7500 units per 10(5) cells per day, protection is completely absent because of impaired generation of tumor-specific cytotoxic T lymphocytes. In comparison, granulocyte-macrophage colony-stimulating factor as immunomodulator induces substantial immunization even at a moderate level of secretion and protects all animals at the maximal obtainable level of secretion. Our findings demonstrate the importance of the interleukin 2 level produced by genetically modified tumor cells and may have substantial impact for the clinical application of cancer vaccines.


Assuntos
Interleucina-2/biossíntese , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Vacinas , Animais , Relação Dose-Resposta a Droga , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Imunização , Esquemas de Imunização , Camundongos , Camundongos Endogâmicos DBA , Plasmídeos , Mapeamento por Restrição , Transfecção
16.
Int J Immunopharmacol ; 17(2): 65-73, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7657408

RESUMO

We have previously demonstrated the general usefulness of the adenovirus-enhanced transferrinfection (AVET) in the generation of IL-2 producing tumor vaccines. By optimizing different parameters of the transfection protocol we were able to transform the poorly immunogenic M-3 mouse melanoma cell line into a potent immunogen. A long-lasting immunity was demonstrated after administration of the IL-2 releasing vaccine, since immunized animals successfully rejected native M-3 melanoma cells even after a period of more than 6 months. We also demonstrated that in vivo administration of such a vaccine is safe since transmission of the transfected IL-2 gene in host organs was not detected. IL-2 production ceased 2 days after injection because the engineered cells were destroyed. However, RT-PCR analysis of the site of vaccine injection suggests that IL-2 exerts its effects not only directly but also by inducing a set of other immunomodulator cytokines in situ that are probably indispensable in inducing a host response. We conclude that AVET of IL-2 into tumor cells is a safe and efficient method for the generation of tumor vaccines.


Assuntos
Interleucina-2/imunologia , Melanoma Experimental/prevenção & controle , Vacinação , Adenoviridae/genética , Animais , Feminino , Interleucina-2/biossíntese , Interleucina-2/genética , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos DBA , Fatores de Tempo , Transfecção/métodos , Células Tumorais Cultivadas , Vacinas Atenuadas/farmacologia
17.
Virology ; 205(1): 254-61, 1994 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-7975222

RESUMO

Adenovirus entry into its host cell transiently permeabilizes the cell allowing the coentry of reagents such as DNA. We compare here adenovirus inactivation with beta-propiolactone and several psoralen derivatives, seeking reagents that disrupt the viral genome without impairing the viral entry functions. No virus replication can be detected after 8-methoxypsoralen (8-MOP) modification. Viral transcription is not detectable by Northern analysis, and reverse transcriptase/PCR analysis demonstrates at least a 1000-fold decrease in viral transcription after 8-MOP treatment. Using [3H]8-MOP, the psoralen is found to enter the virus capsid and react throughout the viral genome, with approximately one psoralen modification per 100 bp of viral DNA. This inactivated adenovirus allows us to deliver DNA to target cells without interference from adenovirus gene expression or replication. Furthermore, we can now study the host cell response to adenovirus entry without the complications of adenovirus gene expression.


Assuntos
Adenoviridae/efeitos dos fármacos , Capsídeo/fisiologia , Metoxaleno/farmacologia , Transcrição Gênica , Replicação Viral/efeitos dos fármacos , Adenoviridae/genética , Adenoviridae/fisiologia , Sequência de Bases , Linhagem Celular , Efeito Citopatogênico Viral , Primers do DNA , Regulação Viral da Expressão Gênica , Genoma Viral , Humanos , Fusão de Membrana , Dados de Sequência Molecular , Propiolactona/farmacologia , Ensaio de Placa Viral
18.
Glycobiology ; 4(4): 429-35, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7827404

RESUMO

The mucin-type carbohydrate Tn cryptantigen (GalNAc alpha 1-O-Ser/Thr, where GalNAc is N-acetyl-D-galactosamine) is expressed in many carcinomas, in haemopoietic disorders including the Tn syndrome, and on human immunodeficiency virus (HIV) coat glycoproteins, but is not expressed on normal, differentiated cells because of the expression of a Tn-processing galactosyltransferase. Using Jurkat T leukaemic cells which express high levels of Tn antigen due to deficient Tn galactosylation, we have established the Tn antigen-mediated gene transfer and demonstrate the considerable efficiency of this approach. We used poly(L-lysine) conjugates of the monoclonal antibody 1E3 directed against the Tn antigen to deliver the luciferase and beta-galactosidase reporter genes to Jurkat cells by receptor-mediated endocytosis. Addition of unconjugated 1E3 reduced transfection efficiency in a concentration-dependent manner and incubation with free GalNAc abolished DNA transfer completely, indicating that gene delivery is indeed mediated by the Tn antigen. Pre-treatment of Jurkat cells with Vibrio cholerae sialidase, which uncovers additional Tn antigens, resulted in an improvement of gene transfection. Both human and chicken adenovirus particles attached to the DNA/polylysine complex strongly augmented transgene expression. When the beta-galactosidase (lacZ) gene was delivered to Jurkat cells by Tn-mediated endocytosis, up to 60% of the cells were positive in the cytochemical stain using 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside (X-gal) as a chromogenic substrate. The efficiency of the transferrin receptor-mediated DNA uptake into Jurkat cells was comparatively low, although these cells were shown to express considerable amounts of transferrin receptor. We show here that a mucin-type carbohydrate antigen mediates highly efficient DNA uptake by endocytosis into Jurkat T cells. This method represents a 50-fold improvement of Jurkat cell transfection efficiency over other physical gene transfer techniques. Specific gene delivery to primary cancer cells exhibiting Tn epitopes may especially be desirable in immunotherapy protocols.


Assuntos
Antígenos Glicosídicos Associados a Tumores/genética , Técnicas de Transferência de Genes , Leucemia de Células T/genética , Adenoviridae/genética , Animais , Anticorpos Monoclonais , Antígenos Glicosídicos Associados a Tumores/metabolismo , DNA/genética , Expressão Gênica , Genes Reporter , Terapia Genética , Vetores Genéticos , Humanos , Óperon Lac , Células Tumorais Cultivadas/metabolismo
19.
Proc Natl Acad Sci U S A ; 91(11): 5148-52, 1994 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-8197198

RESUMO

Hemophilia A is caused by defects in the factor VIII gene. This results in life-threatening hemorrhages and severe arthropathies. Today, hemophiliacs are treated with human blood-derived factor VIII. In the future, it may be possible to use gene therapy to avoid long-term complications of conventional therapy and to improve the quality of life. However, initial gene therapy models using retroviral vectors and nonviral gene transfer techniques to introduce factor VIII gene constructs have been hampered by low expression levels of factor VIII. We show here that high expression levels of the B-domain-deleted human factor VIII in primary mouse fibroblasts and myoblasts are obtained by using receptor-mediated, adenovirus-augmented gene delivery (transferrinfection). We demonstrate that, presumably owing to the high molecular weight of factor VIII or its metabolic instability, secretion into the blood and attainment of therapeutic in vivo levels of factor VIII is achieved only if transfected autologous primary fibroblasts or myoblasts are delivered to the liver or spleen, but not if myoblasts are implanted into muscle, a strategy known to be successful for factor IX delivery.


Assuntos
Adenoviridae/genética , Fator VIII/biossíntese , Fibroblastos/transplante , Baço/metabolismo , Animais , Células Cultivadas , Fator VIII/genética , Fibroblastos/citologia , Humanos , Camundongos , Músculos/citologia , Transfecção , Transferrina
20.
J Biol Chem ; 269(18): 13551-7, 1994 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-8175790

RESUMO

Stimulation of endothelial cells by cytokines and bacterial lipopolysaccharide leads to activation of the transcription factor NF-kappa B. NF-kappa B in turn regulates the expression of several genes involved in the inflammatory reaction, including cell adhesion molecules, interleukins, and transcription factors. One of these induced genes encodes an inhibitor of NF-kappa B, ECI-6/I kappa B alpha, that contains in its 5' regulatory region six consensus binding sites for NF-kappa B. We demonstrate here that these sites display striking differences in their ability in vitro to bind to various NF-kappa B subunits. In vivo, all six sites contribute, though to varying degrees, to transcription from the ECI-6/I kappa B alpha promoter, as demonstrated by deletion and mutation analysis. Among the NF-kappa B subunits tested p65, the p65/p50 heterodimer and, to a lesser extent, c-Rel, are able to activate transcription, whereas p50 or p50/Re1B were inactive. Since many genes regulated by NF-kappa B contain only one or two DNA-binding sites for this transcription factor, the presence of six functional NF-kappa B-binding sites in the ECI-6/I kappa B alpha promoter represents a unique feature of this gene.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas I-kappa B , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Lipopolissacarídeos/farmacologia , Dados de Sequência Molecular , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Oligodesoxirribonucleotídeos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-rel , Deleção de Sequência , Suínos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...