Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Cancer Gene Ther ; 19(11): 757-66, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956041

RESUMO

Sindbis viral vectors target and inhibit the growth of various solid tumors in mouse models. However, their efficacy against blood cancer has not been well established. Here, we show that Sindbis vectors infect and efficiently trigger apoptosis in mouse BW5147 malignant hematopoietic T-cells, but only at low levels in human lymphoma and leukemia cells (Jurkat, Karpas, CEM, DHL and JB). The Mr 37/67 kD laminin receptor (LAMR) has been suggested to be the receptor for Sindbis virus. However, JB cells, which are infected by Sindbis at low efficiency, express high levels of LAMR, revealing that additional factors are involved in Sindbis tropism. To test the infectivity and therapeutic efficacy of Sindbis vectors against malignant hematopoietic cells in vivo, we injected BW5147 cells intraperitoneally into (C3HXAKR) F1 hybrid mice. We found that Sindbis vectors targeted the tumors and significantly prolonged survival of tumor-bearing mice. We also tested the Sindbis vectors in a transgenic CD4-Rgr model, which spontaneously develop thymic lymphomas. However, infectivity in this model was less efficient. Taken together, these results demonstrate that Sindbis vectors have the potential to target and kill hematopoietic malignancies in mice, but further research is needed to evaluate the mechanism underlining the susceptibility of human lymphoid malignancies to Sindbis therapy.


Assuntos
Vetores Genéticos/metabolismo , Neoplasias Hematológicas/metabolismo , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Sindbis virus/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Apoptose , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Neoplasias Hematológicas/terapia , Humanos , Injeções Intraperitoneais , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Metástase Neoplásica , Receptores de Laminina/genética , Receptores de Laminina/metabolismo , Sindbis virus/genética , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia
2.
Cancer Gene Ther ; 19(8): 588-91, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22677940

RESUMO

Oncolytic viruses (OVs) have shown great anti-cancer potential in animal models, but only modest success in early clinical trials. A better understanding of the mechanisms underlining OV efficacy is needed to resolve this discrepancy. In the clinic, OV therapy will likely be combined with traditional chemotherapy, underscoring the need to also evaluate the interactions between these therapeutic modalities. Here we show that combining Sindbis viral vector therapy with the topoisomerase inhibitor irinotecan (CPT-11) results in the long-term survival of about 35% of SCID mice bearing aggressively growing ES2 human ovarian cancer. Single-agent treatments did not result in long-term survival. Flow cytometry analysis, bioluminescent imaging and survival experiments revealed that Sindbis and CPT-11 utilize non-overlapping natural killer (NK)-cell-dependent and -independent anti-cancer mechanisms, respectively. Notably, the combinatorial therapy was only effective in the presence of NK cells. These results highlight the hidden role of immune cell activation in combinatorial cancer therapy involving OVs and provide a potential method for tackling tumor cell resistance to cancer therapy while limiting treatment-related side effects.


Assuntos
Células Matadoras Naturais , Neoplasias Experimentais , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Sindbis virus/genética , Animais , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Feminino , Humanos , Irinotecano , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Camundongos , Camundongos SCID , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/virologia , Neoplasias Ovarianas/terapia , Inibidores da Topoisomerase/administração & dosagem
3.
Oncogene ; 30(12): 1481-8, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21102518

RESUMO

Targeting oncogenic microRNAs (miRNAs) is emerging as a promising strategy for cancer therapy. In this study, we provide proof of principle for the safety and efficacy of miRNA targeting against metastatic tumors. We tested the impact of targeting miR-182, a pro-metastatic miRNA frequently overexpressed in melanoma, the in vitro silencing of which represses invasion and induces apoptosis. Specifically, we assessed the effect of anti-miR-182 oligonucleotides synthesized with 2' sugar modifications and a phosphorothioate backbone in a mouse model of melanoma liver metastasis. Luciferase imaging showed that mice treated with anti-miR-182 had a lower burden of liver metastases compared with control. We confirmed that miR-182 levels were effectively downregulated in the tumors of anti-miR-treated mice compared with tumors of control-treated mice, both in the liver and in the spleen. This effect was accompanied by an upregulation of multiple miR-182 direct targets. Transcriptional profiling of tumors treated with anti-miR-182 or with control oligonucleotides revealed an enrichment of genes controlling survival, adhesion and migration modulated in response to anti-miR-182 treatment. These data indicate that in vivo administration of anti-miRs allows for efficient miRNA targeting and concomitant upregulation of miRNA-controlled genes. Our results demonstrate that the use of anti-miR-182 is a promising therapeutic strategy for metastatic melanoma and provide a solid basis for testing similar strategies in human metastatic tumors.


Assuntos
Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Melanoma/secundário , Melanoma/terapia , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos Antissenso/uso terapêutico , Neoplasias Cutâneas/patologia , Animais , Humanos , Camundongos , MicroRNAs/genética , Oligonucleotídeos Antissenso/genética , Carga Tumoral
4.
Cancer Gene Ther ; 17(4): 244-55, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19798121

RESUMO

Genetic instability of cancer cells generates resistance after initial responses to chemotherapeutic agents. Several oncolytic viruses have been designed to exploit specific signatures of cancer cells, such as important surface markers or pivotal signaling pathways for selective replication. It is less likely for cancer cells to develop resistance given that mutations in these cancer signatures would negatively impact tumor growth and survival. However, as oncolytic viral vectors are large particles, they suffer from inefficient extravasation from tumor blood vessels. Their ability to reach cancer cells is an important consideration in achieving specific oncolytic targeting and potential vector replication. Our previous studies indicated that the Sindbis viral vectors target tumor cells by the laminin receptor. Here, we present evidence that modulating tumor vascular leakiness, using VEGF and/or metronomic chemotherapy regimens, significantly enhances tumor vascular permeability and directly enhances oncolytic Sindbis vector targeting in tumor models. Because host-derived vascular endothelium cells are genetically stable and less likely to develop resistance to chemotherapeutics, a combined metronomic chemotherapeutics and oncolytic vector regimen should provide a new approach for cancer therapy. This mechanism could explain the synergistic treatment outcomes observed in clinical trials of combined therapies.


Assuntos
Infecções por Alphavirus/terapia , Vetores Genéticos , Neovascularização Patológica/prevenção & controle , Terapia Viral Oncolítica , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/terapia , Sindbis virus/fisiologia , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Western Blotting , Permeabilidade da Membrana Celular , Terapia Combinada , Cricetinae , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Camundongos SCID , Neuroblastoma/irrigação sanguínea , Neuroblastoma/terapia , Neuroblastoma/virologia , Neoplasias Ovarianas/virologia , Paclitaxel/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cell Death Dis ; 1: e42, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21243100

RESUMO

The 37/67 kDa laminin receptor (LAMR) is a multifunctional protein, acting as an extracellular receptor, localizing to the nucleus, and playing roles in rRNA processing and ribosome assembly. LAMR is important for cell viability; however, it is unclear which of its functions are essential. We developed a silent mutant LAMR construct, resistant to siRNA, to rescue the phenotypic effects of knocking down endogenous LAMR, which include inhibition of protein synthesis, cell cycle arrest, and apoptosis. In addition, we generated a C-terminal-truncated silent mutant LAMR construct structurally homologous to the Archaeoglobus fulgidus S2 ribosomal protein and missing the C-terminal 75 residues of LAMR, which displays more sequence divergence. We found that HT1080 cells stably expressing either silent mutant LAMR construct still undergo arrest in the G1 phase of the cell cycle when treated with siRNA. However, the expression of full-length silent mutant LAMR rescues cell viability, whereas the expression of the C-terminal-truncated LAMR does not. Interestingly, we also found that both silent mutant constructs restore protein translation and localize to the nucleus. Our findings indicate that the ability of LAMR to regulate viability is associated with its C-terminal 75 residues. Furthermore, this function is distinct from its role in cell proliferation, independent of its ribosomal functions, and may be regulated by a nonnuclear localization.


Assuntos
Receptores de Laminina/química , Receptores de Laminina/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Sequência de Aminoácidos , Aminoácidos , Sequência de Bases , Linhagem Celular Tumoral , Núcleo Celular , Sobrevivência Celular , Fase G1 , Técnicas de Silenciamento de Genes , Humanos , Dados de Sequência Molecular , Peso Molecular , Mutação/genética , Fenótipo , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Relação Estrutura-Atividade
6.
Gene Ther ; 16(2): 291-6, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18818670

RESUMO

A major concern of using viral gene therapy is the potential for uncontrolled vector propagation and infection that might result in serious deleterious effects. To enhance the safety, several viral vectors, including vectors based on Sindbis virus, were engineered to lose their capability to replicate and spread after transduction of target cells. Such designs, however, could dramatically reduce the therapeutic potency of the viral vectors, resulting in the need for multiple dosages to achieve treatment goals. Earlier, we showed that a replication-defective (RD) Sindbis vector achieved specific tumor targeting without any adverse effects in vivo. Here, we present a replication-competent Sindbis viral vector that has an hsvtk suicide gene incorporated into ns3, an indispensable non-structural gene for viral survival. The capability of viral propagation significantly increases tumor-specific infection and enhances growth suppression of tumor compared with the conventional RD vectors. Furthermore, in the presence of the prodrug ganciclovir, the hsvtk suicide gene serves as a safety mechanism to prevent uncontrolled vector propagation. In addition to suppressing vector propagation, toxic metabolites, generated by prodrug activation, could spread to neighboring uninfected tumor cells to further enhance tumor killing.


Assuntos
Genes Transgênicos Suicidas , Terapia Genética/métodos , Vetores Genéticos/genética , Neoplasias Experimentais/terapia , Sindbis virus/genética , Animais , Antivirais/farmacologia , Ganciclovir/farmacologia , Marcação de Genes/métodos , Camundongos , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Pró-Fármacos/farmacologia , Sindbis virus/fisiologia , Replicação Viral/genética
7.
Gene Ther ; 14(15): 1166-74, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17508007

RESUMO

Our previous studies suggest that replication-defective Sindbis vectors might be promising agents for specific tumor targeting and detection. However, the effects of innate and/or adaptive anti-viral immunity, in particular, the IFN-I/STAT1 signaling pathway, may impact their therapeutic potential. Using a bioluminescent imaging system, we demonstrate that although most normal cells are not permissively transduced by replication-defective Sindbis vector, transduction of liver non-sinusoidal endothelial occurs the first time IFN-I/STAT1 signaling deficient mice are inoculated with the vector. Transduction of some cells is not surprising since STAT1 knockout animals show significant delay in IFN responses such as the production of IFN-alpha/beta and transcriptional activation of several anti-viral genes (IRF7, RIG-I, PKR, TLR3, USP18, ISG15). However, beyond the initial vector transduction, which resolves rapidly, secondary inoculums of Sindbis vectors do not transduce any liver cells, suggesting that an alternative antiviral pathway may protect against further transduction. Other known signaling pathways were examined using mice lacking functional TLR3, tumor necrosis factoralphaR or nuclear factor-kappa B (p50). Surprisingly, none of those pathways seem to play a significant role in anti-Sindbis responses. Thus it appears that in vivo, in contrast to the ready transduction of tumor cells, transduction of normal cells by replication-defective Sindbis vector is limited, possibly by a novel mechanism.


Assuntos
Infecções por Alphavirus/imunologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Interferon Tipo I/genética , Neoplasias/terapia , Sindbis virus/genética , Animais , Vetores Genéticos/imunologia , Imunidade , Imunidade Inata , Interferon Tipo I/imunologia , Fígado/imunologia , Fígado/virologia , Luminescência , Camundongos , Camundongos Knockout , NF-kappa B/genética , Neoplasias/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Receptor 3 Toll-Like/genética , Transdução Genética/métodos
8.
DNA Cell Biol ; 20(3): 141-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11313017

RESUMO

Deltex is a component of the Notch signaling network, which mediates cellular differentiation, proliferation, and apoptosis during development. Murine Deltex was initially isolated as a cDNA transcript that displayed increased expression in T-cell tumors induced by gamma irradiation. The in vivo function of Deltex is unknown; however, the emerging role of Notch signaling in T-cell development and lymphomagenesis indirectly supports a role for Deltex in these processes. To investigate the regulation of Deltex expression in both normal and transformed tissue, we have begun analyzing the Deltex genomic locus. Here, we report the exon-intron organization of Deltex and map the locus to the middistal region of mouse chromosome 5, tightly linked to the Adam1a, Lnk, Tbx5, and Nos1 loci. The human homolog of Deltex has been localized to chromosome 12.


Assuntos
Proteínas de Transporte , Mapeamento Cromossômico , Cromossomos Humanos Par 12 , Proteínas/genética , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Proteínas ADAM , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Clonagem Molecular , Éxons , Fertilinas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Íntrons , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo I , Proteínas/metabolismo , Receptores Notch , Homologia de Sequência de Aminoácidos , Proteínas com Domínio T/genética
9.
DNA Cell Biol ; 19(7): 383-8, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10945228

RESUMO

New modalities of treatment for small-cell lung cancer (SCLC) are needed, because the majority of patients continue to die of disseminated disease despite an initial response to conventional chemotherapy. Abnormal surface expression of the neural-cell adhesion molecule (NCAM) has been noted to be highly associated with SCLC. We examined the ability and efficiency of a streptavidin-Protein A (ST-PA) fusion protein complexed with an anti-NCAM monoclonal antibody (Mab) to transfer biotinylated beta-galactosidase into human SCLC cell lines NCI-H69, NCI-H526, and NCI-H446. When the surface molecule NCAM was targeted with this system, more than 99% of the targeted cells internalized and exhibited beta-galactosidase activity. In addition, we evaluated cytotoxic activity against SCLC lines NCI-H69 and NCI-H526 by efficient delivery of biotinylated glucose oxidase using the same ST-PA/anti-NCAM Mab complex. Cytotoxicity of the transduced cells (SCLC) was 10-fold and 100-fold greater, respectively, than the glucose oxidase control. This system could be widely applied for specific therapy of cancer cells by targeting unique surface molecules (antigens) using the corresponding Mab/ST-PA complex to transfer a variety of effector molecules; e.g., immunotoxic compounds, into target cells with a high degree of efficiency and specificity.


Assuntos
Carcinoma de Células Pequenas/genética , Técnicas de Transferência de Genes , Neoplasias Pulmonares/genética , Estreptavidina/genética , Toxinas Biológicas/genética , Animais , Anticorpos Monoclonais/genética , Biotina/genética , Carcinoma de Células Pequenas/patologia , Carcinoma de Células Pequenas/terapia , Contagem de Células , Sobrevivência Celular , Glucose Oxidase/genética , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos SCID , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/imunologia , Proteínas Recombinantes de Fusão/genética , Transdução Genética , Células Tumorais Cultivadas , beta-Galactosidase/genética
10.
Int Immunol ; 12(4): 479-86, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10744649

RESUMO

The cytotoxicity reaction of murine CD8 T lymphocytes has been found to be strongly inhibited by nanomolar concentrations of hypericin, a lipophilic dianthraquinone with photodynamic properties. Cytotoxic T lymphocyte (CTL)-induced target cell apoptosis, as well as exocytosis of cytolytic granules from these cells, were ablated by hypericin, administered at the onset of the reaction, without affecting CTL viability. The inhibition of cytolysis occurred without the light irradiation which is essential for photosensitization. The findings suggest that the action of hypericin targets the effector CTL; however, apoptosis induced in murine L-cells with recombinant tumor necrosis factor (TNF)-alpha was also prevented by hypericin. Since hypericin is a known inhibitor of protein kinase C, MAP kinase and at least one other tyrosine kinase, this inhibitory activity could play a role in the down-modulation of CTL-induced cytotoxicity. Furthermore, our studies show that the action of hypericin induces rapid dephosphorylation of phospholipids associated with low-density membranes in CTL, but not with membranes of the cytotoxic granules. The ability of hypericin to interfere with cytotoxicity may render it useful in the treatment of T cell-mediated diseases.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/imunologia , Imunossupressores/farmacologia , Perileno/análogos & derivados , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Animais , Antracenos , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/imunologia , Testes Imunológicos de Citotoxicidade , Exocitose/efeitos dos fármacos , Exocitose/imunologia , Imunossupressores/uso terapêutico , Ligantes , Luz , Camundongos , Perileno/farmacologia , Perileno/uso terapêutico , Fosfolipídeos/metabolismo , Fosforilação/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/metabolismo
11.
Int Immunol ; 12(2): 161-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10653851

RESUMO

The transcription of the MHC class I genes is regulated by interaction of cis-elements, located in the 5' genomic flanking regions, with sequence-specific trans-factors. We have identified a cis-regulatory element, 5'-TGACGCG-3', of the H-2D(d) gene. This cyclic adenosine-3',5'-monophosphate regulatory element (CRE)-like sequence, named H-2 binding factor 1 (H-2 BF1) binding motif, is highly conserved among species. In addition, we found that homo- and heterodimers of activation transcription factor 1 (ATF-1) and CRE binding protein (CREB) associate with the H-2 BF1 binding motif and activate transcription of the H-2D(d) gene. Here we demonstrate that a homologue of ATF-1, originally isolated and designated ATF-1DN, acts as a dominant repressor, blocking the ability of wild-type ATF-1 and CREB to bind to the H-2 BF1 probe in electrophoretic mobility shift assays (EMSA). We have utilized this molecule to analyze the participation of the H-2 BF1 complexes, consisting of the H-2 BF1 binding motif and ATF-1/CREB trans-factors, in the physiological regulation of MHC class I expression in tissue culture cells. A human epidermoid carcinoma cell line, A431, was transfected with ATF-1DN and clones expressing the gene transcripts were selected. When analyzed in the EMSA, nuclear proteins prepared from these clones exhibited a decreased shift of the H-2 BF1 probe corresponding to the levels of the ATF-1DN gene expression. Additionally, MHC class I expression of cells with reduced H-2 BF1 activity was significantly higher than in control cells lacking ATF-1DN. These findings indicate that in these carcinoma cells, the H-2 BF1 complexes negatively regulate the constitutive expression of MHC class I.


Assuntos
Membrana Celular/imunologia , Proteínas de Ligação a DNA/metabolismo , Genes MHC Classe I , Antígenos H-2/metabolismo , Fatores de Transcrição/genética , Transfecção , Fator 1 Ativador da Transcrição , Animais , Clonagem Molecular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Antígeno de Histocompatibilidade H-2D , Humanos , Camundongos , Neomicina/farmacologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
12.
Photochem Photobiol ; 71(2): 188-95, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10687393

RESUMO

Photodynamically induced virus inactivation appears promising in preventing transmission of enveloped virus infections in transfusible blood products. The potential for utilizing hypericin as a photosensitizer to inactivate key enveloped viruses in packed red cell concentrates (PRC) was evaluated. In addition to inactivating effectively > or = 10(6) TCID50 of human immunodeficiency virus (HIV), inactivation of bovine viral diarrhea virus (BVDV) in PRC was used as a model for hepatitis C virus to overcome the deficiency in reliable experimental systems for hepatitis C virus (HCV) inactivation. BVDV was two orders of magnitude more sensitive to inactivation by hypericin than HIV. As part of the virucidal efficacy analyses, the effects of photosensitization on hemopoietic cell lines carrying quiescent integrated HIV provirus were studied as models for evaluating virus inactivation in latently infected cells. Phorbol ester-induced virus production by these cells was effectively prevented by photosensitization with hypericin. A refinement of the illumination conditions, incorporating a monochromatic sodium light source with an emission spectrum coinciding with the absorption peak of hypericin, was highly virucidal, however, caused unacceptable levels of hemolysis. Red blood cells could be protected from phototoxic cellular damage by complexing hypericin with human serum albumin (albumin-hypericin), but the decrease in hemolysis was at the expense of virucidal efficacy. Thus, excitation of hypericin with a fluorescent source appears to be useful potentially for virus inactivation in PRC.


Assuntos
Antivirais/farmacologia , Vírus da Diarreia Viral Bovina/efeitos dos fármacos , Eritrócitos/virologia , HIV-1/efeitos dos fármacos , Perileno/análogos & derivados , Animais , Antracenos , Fármacos Anti-HIV/farmacologia , Bovinos , Sistema Livre de Células , Vírus da Diarreia Viral Bovina/efeitos da radiação , HIV-1/efeitos da radiação , Humanos , Luz , Perileno/farmacologia , Radiossensibilizantes/farmacologia
13.
DNA Cell Biol ; 18(10): 743-9, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10541433

RESUMO

The previously reported streptavidin-TGFalpha chimeric protein-based delivery system (Ohno and Meruelo, DNA Cell Biol. 15:401-406, 1996) could efficiently transfer protein molecules into A431 cells via the epidermal growth factor (EGF) receptor. We have modified this delivery system for the transfer of DNA. For this purpose, we have linked the chimeric protein ST-TGFalpha to DNA through biotinylated polylysine molecules. We show with this system, in the presence of the endosome-destabilizing reagent chloroquine, an average of 50-fold increase in reporter gene expression in comparison with polylysine DNA complexes alone. This gene expression is specific for EGF receptor-expressing cells and is blocked by EGF-binding molecules. These results suggest that the ST-TGFalpha biotinylated polylysine system could be used to deliver DNA to targeted cells.


Assuntos
Endocitose , Receptores ErbB/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Estreptavidina/farmacologia , Transfecção/métodos , Fator de Crescimento Transformador alfa/farmacologia , Animais , Biotinilação , Células COS , Carcinoma de Células Escamosas/patologia , Linhagem Celular Transformada , Chlorocebus aethiops , Cloroquina/farmacologia , DNA Recombinante/administração & dosagem , Portadores de Fármacos , Endossomos/efeitos dos fármacos , Genes Reporter , Humanos , Luciferases/biossíntese , Luciferases/genética , Polilisina/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Vírus 40 dos Símios , Estreptavidina/genética , Fator de Crescimento Transformador alfa/genética , Células Tumorais Cultivadas
14.
Mol Genet Metab ; 67(1): 36-42, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10329021

RESUMO

Sindbis virus has been recognized as a potentially useful virus vector for gene therapy. In an effort to improve its utility and provide cell-targeting capability to gene therapy vectors, we recently developed Sindbis virus vectors possessing chimeric envelopes with cell-specific targeting ability [K. Ohno et al. Nature Biotechnol 15:763-767, 1997; K. Sawai et al. Biochem Biophys Res Commun 248:315-323, 1998]. However, a residual problem associated with Sindbis virus vectors is the apoptotic effect of this virus on infected cells. To address this issue, we have studied the possible role of bcl-2 expression. Bcl-2 expression has been postulated to facilitate the establishment of persistent Sindbis viral infection by blocking virus-induced apoptosis. In this study we produced a Sindbis virus vector capable of expressing human bcl-2 and the reporter gene, lacZ. This chimeric virus (SinRep/lacZ/bcl-2/DH-BB) showed a marked reduction in induced apoptosis in infected cells. For example, after infection with this vector, cell proliferation of BHK cells was 55% of that of uninfected cells 2 days after infection and 40% 3 days after infection. While this reflected a significant degree of apoptosis, the effect was much less pronounced than that seen with wild-type Sindbis virus. Cell proliferation was reduced to 26% 2 days after wild-type virus infection of BHK cells and to only 7% 3 days after infection. Although additional work will be required to eliminate apoptosis induced by Sindbis virus vectors, the studies reported here suggest that such a goal may be achievable after additional modification of the vectors.


Assuntos
Apoptose , Vetores Genéticos , Sindbis virus/genética , Animais , Divisão Celular , Linhagem Celular , Cricetinae , Genes Reporter , Immunoblotting , Rim/metabolismo , Modelos Genéticos , Plasmídeos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção , beta-Galactosidase/metabolismo
15.
Int J Cancer ; 80(1): 110-8, 1999 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-9935240

RESUMO

Transfer of the herpes simplex virus type I thymidine kinase (HSV-TK) gene into tumor cells using virus-based vectors in conjunction with ganciclovir (GCV) exposure provides a potential gene therapy strategy for the treatment of cancer. The possibility of using a novel targetable Sindbis virus expression vector containing the HSV-TK gene was examined. Baby hamster kidney (BHK) cells and several human tumor cells infected with a Sindbis virus containing the HSV-TK gene showed strong expression of HSV-TK protein. Cells transduced with the HSV-TK gene exhibited increased TK activity, ranging from 3- to 20-fold over an average baseline level. The human HeLa-CD4+ cells infected with recombinant Sindbis virus containing the HSV-TK gene were sensitive to low concentrations of GCV (0.1-1 microg/ml) and the 50% growth inhibitory concentration (IC50) was 0.6 microg/ml. We also demonstrated applications of cell type-specific Sindbis virus-mediated antigen-antibody targeting of the HSV-TK/GCV system in vitro. Sindbis virus containing the HSV-TK gene packaged in a helper virus displaying the IgG-binding domain of protein A on its envelope could infect various tumor cell lines in the presence of specific antibodies that recognize antigens on their surfaces. HSV-TK-transduced tumor cell lines exhibited sensitivity to GCV. Our data suggest the potential for targeted gene therapy of the HSV-TK/GCV system using a cell type-specific recombinant Sindbis virus vector-antibody system.


Assuntos
Antivirais/toxicidade , Ganciclovir/toxicidade , Terapia Genética/métodos , Sindbis virus/genética , Timidina Quinase/genética , Transfecção/métodos , Animais , Antivirais/uso terapêutico , Sequência de Bases , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Ganciclovir/uso terapêutico , Vetores Genéticos , Células HeLa , Humanos , Rim , Dados de Sequência Molecular , Recombinação Genética , Mapeamento por Restrição , Timidina Quinase/antagonistas & inibidores , Células Tumorais Cultivadas
16.
Br J Cancer ; 79(3-4): 423-32, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10027308

RESUMO

The mechanism of cell death induction by dimethyl tetrahydroxyhelianthrone (DTHe), a new second-generation photodynamic sensitizer, is analysed in human leukaemic cell lines in comparison with the structurally related hypericin. DTHe has a broad range of light spectrum absorption that enables effective utilization of polychromatic light. Photosensitization of HL-60 cells with low doses of DTHe (0.65 microM DTHe and 7.2 J cm(-2) light energy) induced rapid apoptosis of > or =90% of the cells. At doses > or =2 microM, dying cells assumed morphological necrosis with perinucleolar condensation of chromatin in HL-60 and K-562 cell lines. Although nuclear fragmentation that is characteristic to apoptosis was prevented, DNA digestion to oligonucleosomes proceeded unhindered. Such incomplete apoptosis was more prevalent with the related analogue hypericin throughout most doses of photosensitization. Despite hypericin being a stronger photosensitizer, DTHe exhibited advantageous phototoxic properties to tumour cells, initiating apoptosis at concentrations about threefold lower than hypericin. Photosensitization of the cells induced dissociation of the nuclear envelope, releasing lamins into the cytosol. DTHe also differed from hypericin in effects exerted on the nuclear lamina, causing release of an 86-kDa lamin protein into the cytosol that was unique to DTHe. Within the nucleus, nuclear envelope lamin B underwent covalent polymerization, which did not affect apoptotic nuclear fragmentation at low doses of DTHe. At higher doses, polymerization may have been extensive enough to prevent nuclear collapse. Hut-78, CD4+ cells were resistant to the photodynamically activated apoptotic pathway. Beyond the tolerated levels of photodynamic damage, these cells died exclusively via necrosis. Hut-78 cells overexpress Bcl-X(L) as well as a truncated Bcl-X(L)tr isoform that could contribute to the observed resistance to apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Perileno/análogos & derivados , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2 , Radiossensibilizantes/farmacologia , Antracenos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , DNA de Neoplasias/genética , Relação Dose-Resposta a Droga , Genes bcl-1/efeitos dos fármacos , Genes bcl-2/efeitos dos fármacos , Células HL-60/efeitos dos fármacos , Humanos , Necrose , Perileno/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteína X Associada a bcl-2
17.
Gynecol Oncol ; 72(1): 16-25, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9889024

RESUMO

In gene therapy, the herpes simplex virus thymidine kinase (HSV-tk) gene is widely used as a suicide agent. Tumor cells expressing HSV-tk are sensitive to nucleoside analogs such as ganciclovir (GCV). An advantage of this system is the bystander killing effect whereby HSV-tk-positive cells exposed to GCV are lethal to surrounding HSV-tk-negative cells. We transfected the HSV-tk gene into a human cervical adenocarcinoma cell line, BU25TK-, and a human endometrial adenocarcinoma cell line, HHUA, by the Lipofectine method. The sensitivity of HSV-tk-positive cells to GCV and bystander killing effect on HSV-tk-negative cells were examined in vitro. HSV-tk-positive cells were sensitive to GCV at concentrations of 1 to 100 microg/ml in a dose- and time-dependent manner. The growth of HSV-tk-negative cells was inhibited when the population of cultured cells contained more than about 3% HSV-tk-positive cells. Moreover, for BU25TK- cells, HSV-tk-positive cells were injected into SCID mice subcutaneously and the effects of GCV therapy and bystander killing at a daily concentration of 25 mg/kg for 14 days were examined. HSV-tk-positive tumors transduced into SCID mice almost disappeared upon GCV treatment. Furthermore, tumor reduction was observed when mixtures of HSV-tk-negative cells containing more than 20% HSV-tk-positive cells were injected into SCID mice. In conclusion, the HSV-tk/GCV system might be applied to both cervical and endometrial adenocarcinoma.


Assuntos
Adenocarcinoma/terapia , Terapia Genética/métodos , Simplexvirus , Timidina Quinase , Neoplasias Uterinas/terapia , Adenocarcinoma/genética , Animais , Feminino , Ganciclovir/uso terapêutico , Humanos , Camundongos , Camundongos SCID , Simplexvirus/genética , Timidina Quinase/genética , Células Tumorais Cultivadas , Neoplasias Uterinas/genética
18.
Mol Genet Metab ; 64(1): 44-51, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9682217

RESUMO

In this study, we developed a cell-specific mRNA transfection system using streptavidin-protein A (ST-PA) fusion protein and monoclonal antibodies (mAbs). We previously reported that ST-PA fusion protein and mAb complexes can transfer certain biotinylated proteins into specific cell types. At this time, we combined an in vitro transcribed biotinylated and self-replicating Sindbis virus genomic RNA with ST-PA fusion protein and mAbs. In the presence of cationic liposomes, to prevent RNA degradation, this complex is able to transfect a reporter gene to specific cancer cells in a mAb does-dependent manner. Even in the absence of cationic liposomes, biotinylated mRNA, ST-PA fusion, and mAb complexes can transfer some types of cancer cell suspension cultures. This cell-specific transfection system is a novel method of introducing various mRNAs into cells that results in high levels of transient protein expression.


Assuntos
RNA Mensageiro/genética , Transfecção/métodos , Animais , Anticorpos Monoclonais , Biotinilação , Antígenos CD4/imunologia , Cátions , Linhagem Celular , Cricetinae , Eletroporação , Receptores ErbB/imunologia , Terapia Genética , Humanos , Imunoglobulina G , Óperon Lac , Lipídeos , Lipossomos , Especificidade de Órgãos , Compostos de Amônio Quaternário , Proteínas Recombinantes de Fusão , Proteína Estafilocócica A , Estreptavidina , Células Tumorais Cultivadas
19.
Biochem Biophys Res Commun ; 248(2): 315-23, 1998 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-9675133

RESUMO

The development of Sindbis virus vectors that can target specific cell types would provide an important gene therapy strategy. We explored the possibility of designing a Sindbis virus vector that can target human choriocarcinoma cells via ligand-receptor interaction. The Sindbis virus envelope gene was modified by insertion of the alpha- and beta-hCG genes. The chimeric helper RNA was then transfected into BHK cells along with a virus-based expression vector, allowing the production of virus particles containing hCG-envelope chimeras. The hCG-envelope chimeric virus vector has minimal infectivities against BHK cells and human cancer cells which do not contain LH/CG receptors on their surface. This vector can, however, infect and transfer a reporter gene to choriocarcinoma cells as well as other cells bearing LH/CG receptors. This chimeric Sindbis virus vector may provide a novel approach for gene therapy of gestational trophoblast disease and placental dysfunction.


Assuntos
Coriocarcinoma/terapia , Gonadotropina Coriônica/genética , Vetores Genéticos/genética , Sindbis virus/genética , Transfecção/genética , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Linhagem Celular , Marcação de Genes/métodos , Terapia Genética/métodos , Humanos , Receptores do LH/genética , Proteínas Recombinantes de Fusão/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética
20.
J Immunol ; 160(12): 5907-14, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9637503

RESUMO

Resistance to radiation leukemia virus (RadLV)-induced leukemia is correlated with an increase in H-2Dd expression on the thymocyte surface. It has been shown that elevated H-2Dd expression on infected thymocytes is a result of elevated mRNA transcription and that the transcriptional increase is correlated with elevated levels of a DNA binding activity, H-2 binding factor 1 (H-2 BF1), which recognizes the 5'-flanking sequence (5'-TGACGCG-3') of the H-2Dd gene. Recently, it has been shown that the activation transcription factor 1 (ATF-1) homodimer is one form of the H-2 BF1 complex. Here we demonstrate that the cAMP response element binding protein (CREB) homodimer and the heterodimer of CREB/ATF-1 also recognize the cis regulatory motif and are two additional forms of the H-2 BF1 complex. The levels of mRNA encoding ATF-1 and CREB were both increased in RadLV-infected thymocytes that showed increased levels of H-2 mRNA. Also, all three H-2 BF1 binding activities, ATF-1 homodimer, CREB homodimer, and ATF-1/CREB heterodimer, were increased in RadLV-infected thymocytes that expressed high levels of H-2Dd Ag on the cell surface. Transfection experiments demonstrated that ATF-1 and CREB activated a reporter plasmid containing the H-2 BF1 motif. These observations strongly suggest that both ATF-1 and CREB are involved in the regulation of H-2 gene expression following RadLV infection of mouse thymocytes.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica , Antígenos H-2/biossíntese , Isoantígenos/biossíntese , Complexo Principal de Histocompatibilidade/imunologia , Fatores de Transcrição/metabolismo , Fator 1 Ativador da Transcrição , Animais , Proteínas de Ligação a DNA/metabolismo , Dimerização , Antígenos H-2/genética , Antígenos H-2/imunologia , Antígeno de Histocompatibilidade H-2D , Isoantígenos/genética , Isoantígenos/imunologia , Leucemia Experimental/genética , Leucemia Experimental/imunologia , Complexo Principal de Histocompatibilidade/genética , Camundongos , Vírus da Leucemia Induzida por Radiação , Infecções por Retroviridae/genética , Infecções por Retroviridae/imunologia , Timoma/genética , Timoma/imunologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA