Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 77(20): 11082-93, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14512556

RESUMO

Herpes simplex virus type 1 (HSV-1) can produce disseminated, systemic infection in neonates and patients with AIDS or other immunocompromising diseases, resulting in significant morbidity and mortality in spite of antiviral therapy. Components of host immunity that normally limit HSV-1 to localized epithelial and neuronal infection remain incompletely defined. We used in vivo bioluminescence imaging to determine effects of type I and II interferons (IFNs) on replication and tropism of HSV-1 infection in mice with genetic deficiency of type I, type II, or both type I and II IFN receptors. Following footpad or ocular infection of mice lacking type I IFN receptors, HSV-1 spread to parenchymal organs, including lung, liver, spleen, and regional lymph nodes, but mice survived. Deletion of type I and II IFN receptors produced quantitatively greatest and most widespread dissemination of virus to visceral organs and the nervous system, and these mice invariably died after ocular or footpad infection. Type II receptor knockout and wild-type mice had comparable viral replication and localization, with no systemic spread of HSV-1 or lethality. Therefore, while isolated deficiency of type II IFN receptors did not affect pathogenesis, loss of these receptors in combination with genetic deletion of type I receptors had a profound effect on susceptibility to HSV-1. These data demonstrate different effects of type I and II IFNs in limiting systemic dissemination of HSV-1 and further validate the use of bioluminescence imaging for studies of viral pathogenesis.


Assuntos
Herpesvirus Humano 1/patogenicidade , Receptores de Interferon/fisiologia , Animais , Córnea/virologia , Luciferases/metabolismo , Medições Luminescentes , Proteínas de Membrana , Camundongos , Receptor de Interferon alfa e beta , Replicação Viral , Receptor de Interferon gama
2.
Nat Med ; 9(7): 969-73, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12819780

RESUMO

The ubiquitin-proteasome pathway is the central mediator of regulated proteolysis in cells, and defects in this pathway are associated with cancer and neurodegenerative diseases. To assess 26S proteasome function in living animals, we developed a ubiquitin-luciferase reporter for bioluminescence imaging. The reporter was degraded rapidly under steady-state conditions and stabilized in a dose- and time-dependent manner in response to proteasome inhibitors. Using bioluminescence imaging after one dose of the chemo-therapeutic proteasome inhibitor bortezomib (PS-341), proteasome function in tumor xenografts was blocked within 30 min and returned to nearly baseline by 46 h. After a 2-week regimen of bortezomib, however, imaging of target tumors showed significantly enhanced proteasome inhibition that no longer returned to baseline. The ubiquitin-luciferase reporter enables repetitive tissue-specific analysis of 26S proteasome activity in vivo and should facilitate development and validation of proteasome inhibitors in mouse models, as well as investigations of the ubiquitin-proteasome pathway in disease pathogenesis.


Assuntos
Acetilcisteína/análogos & derivados , Biologia Molecular/métodos , Peptídeo Hidrolases/análise , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma , Acetilcisteína/farmacologia , Animais , Ácidos Borônicos/farmacologia , Bortezomib , Besouros , Inibidores de Cisteína Proteinase/farmacologia , Genes Reporter , Células HeLa , Humanos , Cinética , Leupeptinas/farmacologia , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes , Masculino , Camundongos , Camundongos Nus , Peptídeo Hidrolases/efeitos dos fármacos , Pirazinas/farmacologia , Transfecção , Ubiquitina/genética , Ubiquitina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cancer Res ; 63(8): 1780-8, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12702563

RESUMO

Protein-protein interactions control essential steps in signal transduction pathways and other intracellular processes, and assembly of protein complexes modulates and responds to the regulatory events that exist in living animals. We have used microPET and fluorescence imaging to detect interactions between p53 tumor suppressor and large T antigen (TAg) of SV40 virus in a tetracycline-inducible two-hybrid system. To additionally validate this molecular imaging technique, we investigated whether expression of the reporter gene, comprised of a mutant thymidine kinase from herpes simplex virus 1 fused to green fluorescent protein could quantify relative differences in amounts of interacting hybrid proteins. In HeLa cells stably transfected with the reporter gene and interacting (p53-TAg) or noninteracting (p53 and polyoma virus coat protein) pairs of proteins, treatment with doxycycline produced time- and dose-dependent increases in expression of hybrid proteins. Proportional increases in amounts of reporter gene were produced only in cells expressing p53 and TAg. In mice bearing xenografts of these stably transfected HeLa cells, amounts of hybrid proteins were regulated with doxycycline. Both microPET imaging and biodistribution studies showed time- and dose-dependent increases in accumulation of the reporter substrate 9-(4-[(18)F]-fluoro-3-hydroxymethylbutyl)guanine only in p53-TAg tumors. Fluorescence microscopy of excised tumors also showed corresponding changes in expression of the fusion reporter gene in response to binding of p53 and TAg. These data demonstrate that the imaging two-hybrid system responds in a proportional fashion to increasing amounts of interacting proteins in vivo.


Assuntos
Aciclovir/análogos & derivados , Antígenos Virais de Tumores/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Aciclovir/farmacocinética , Animais , Antígenos Virais de Tumores/biossíntese , Antígenos Virais de Tumores/genética , Proteínas de Fluorescência Verde , Guanina , Células HeLa , Humanos , Processamento de Imagem Assistida por Computador/métodos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Timidina Quinase/biossíntese , Timidina Quinase/genética , Tomografia Computadorizada de Emissão/métodos , Transfecção , Trítio , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
4.
J Virol ; 76(23): 12149-61, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12414955

RESUMO

Mouse models of herpes simplex virus type 1 (HSV-1) infection provide significant insights into viral and host genes that regulate disease pathogenesis, but conventional methods to determine the full extent of viral spread and replication typically require the sacrifice of infected animals. To develop a noninvasive method for detecting HSV-1 in living mice, we used a strain KOS HSV-1 recombinant that expresses firefly (Photinus pyralis) and Renilla (Renilla reniformis) luciferase reporter proteins and monitored infection with a cooled charge-coupled device camera. Viral infection in mouse footpads, peritoneal cavity, brain, and eyes could be detected by bioluminescence imaging of firefly luciferase. The activity of Renilla luciferase could be imaged after direct administration of substrate to infected eyes but not following the systemic delivery of substrate. The magnitude of bioluminescence from firefly luciferase measured in vivo correlated directly with input titers of recombinant virus used for infection. Treatment of infected mice with valacyclovir, a potent inhibitor of HSV-1 replication, produced dose-dependent decreases in firefly luciferase activity that correlated with changes in viral titers. These data demonstrate that bioluminescence imaging can be used for noninvasive, real-time monitoring of HSV-1 infection and therapy in living mice.


Assuntos
Aciclovir/análogos & derivados , Herpes Simples/etiologia , Herpesvirus Humano 1/patogenicidade , Valina/análogos & derivados , Aciclovir/uso terapêutico , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Feminino , Genes Reporter/efeitos dos fármacos , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/isolamento & purificação , Ceratite Herpética/virologia , Luciferases/genética , Medições Luminescentes , Camundongos , Especificidade de Órgãos , Fatores de Tempo , Valaciclovir , Valina/uso terapêutico
5.
Bioconjug Chem ; 13(6): 1226-37, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12440857

RESUMO

To enable concurrent whole body scintigraphy and direct imaging of subcellular localization of permeation peptides, dual-labeled Tat-peptides useful for both radiometric analysis and fluorescence microscopy are desired for molecular imaging applications. Thus, novel dual-labeled D-Tat-peptides comprising Tat-basic domain (hgrkkrrqrrrgc), C-terminus conjugated with fluorescein-5-maleimide (FM) and N-terminus chelated with [(99m)Tc(CO)(3)] via histidine coordination, were synthesized and characterized. In human Jurkat cells, radiotracer uptake and washout studies revealed concentration-dependent accumulation of the dual-labeled Tat-peptide within cells. Subcellular localization of Tat-peptide was confirmed by fluorescence microscopy using an analogous [Re(CO)(3)] dual-labeled Tat-peptide. As seen with C-terminus single-labeled Tat-peptides, localization to the nucleoli was observed with the dual-labeled Tat-peptide, suggesting that the mechanism of Tat-peptide uptake and localization was not dependent on free peptide termini at either end. In Balb/c mice, biodistribution studies performed with the dual-labeled Tat-peptide showed fluorescence intensity by microscopic analysis that visually confirmed and correlated directly with scintigraphic and radiometric data. Of note, following intravenous administration, little brain penetration of these permeation sequences was observed in vivo. His[(99m)Tc(CO)(3)]-, DTPA[(99m)Tc(CO)(3)]-, and epsilon-lys-gly-cys[(99m)Tc(O)]-labeled Tat-peptides showed significant pharmacokinetic differences in liver and kidney depending on labeling strategy, indicating that Tat-peptide biodistribution can be impacted by the chelation moiety coordinated with (99m)Tc. Thus, we have shown that dual-labeled (99m)Tc-tricarbonyl Tat-peptide-FM conjugates can be conveniently synthesized and enable direct comparison of quantitative radiometric and qualitative fluorescence data both in vitro as well as in vivo.


Assuntos
Fluoresceína/análise , Produtos do Gene tat/análise , Histidina/análise , Microscopia de Fluorescência/métodos , Compostos de Organotecnécio/análise , Fragmentos de Peptídeos/análise , Cintilografia/métodos , Animais , Quelantes , Produtos do Gene tat/síntese química , Produtos do Gene tat/metabolismo , Produtos do Gene tat/farmacocinética , Humanos , Concentração de Íons de Hidrogênio , Células Jurkat , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Ácido Pentético , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacocinética , Transporte Proteico , Compostos Radiofarmacêuticos/análise , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
6.
Proc Natl Acad Sci U S A ; 99(10): 6961-6, 2002 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-11997447

RESUMO

Protein-protein interactions control transcription, cell division, and cell proliferation as well as mediate signal transduction, oncogenic transformation, and regulation of cell death. Although a variety of methods have been used to investigate protein interactions in vitro and in cultured cells, none can analyze these interactions in intact, living animals. To enable noninvasive molecular imaging of protein-protein interactions in vivo by positron-emission tomography and fluorescence imaging, we engineered a fusion reporter gene comprising a mutant herpes simplex virus 1 thymidine kinase and green fluorescent protein for readout of a tetracycline-inducible, two-hybrid system in vivo. By using micro-positron-emission tomography, interactions between p53 tumor suppressor and the large T antigen of simian virus 40 were visualized in tumor xenografts of HeLa cells stably transfected with the imaging constructs. Imaging protein-binding partners in vivo will enable functional proteomics in whole animals and provide a tool for screening compounds targeted to specific protein-protein interactions in living animals.


Assuntos
Antígenos Transformantes de Poliomavirus/metabolismo , Herpesvirus Humano 1/enzimologia , Timidina Quinase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Antígenos Transformantes de Poliomavirus/genética , Genes Reporter , Proteínas de Fluorescência Verde , Células HeLa , Herpesvirus Humano 1/genética , Humanos , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Timidina Quinase/genética , Tomografia Computadorizada de Emissão/métodos , Proteína Supressora de Tumor p53/genética , Técnicas do Sistema de Duplo-Híbrido
7.
Mol Imaging ; 1(2): 65-73, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12920846

RESUMO

Toward the goal of monitoring activity of native mammalian promoters with molecular imaging techniques, we stably transfected DU145 prostate carcinoma cells with a fusion construct of enhanced green fluorescent protein (EGFP) and wild-type herpes simplex virus-1 thymidine kinase (HSV1-TK) as a reporter gene driven by the promoter for human elongation factor 1 alpha (EF-1 alpha-EGFP-TK). Using this model system, expression of EGFP was quantified by flow cytometry and fluorescence microscopy, while the HSV1-TK component of the reporter was quantified with 8-[3H]ganciclovir (8-[3H]GCV). As analyzed by flow cytometry, passage of EGFP-TK-DU145 transfected cells (ETK) in vitro resulted in populations of cells with high and low expression of EGFP over time. High and low ETK cells retained 23-fold and 5-fold more GCV, respectively, than control. While differences in uptake and retention of GCV corresponded to relative expression of the reporter gene in each subpopulation of cells as determined by both flow cytometry (EGFP) and quantitative RT-PCR, the correlation was not linear. Furthermore, in high ETK cells, net retention of various radiolabeled nucleoside analogues varied; the rank order was 8-[3H]GCV < 9-(4-fluoro-3-hydroxymethylbutyl)guanine ([18F]FHBG) approximately 8-[3H]penciclovir (8-[3H]PCV) < 2'-fluoro-2'-deoxy-5-iodouracil-beta-D-arabinofuranoside (2-[14C]FIAU). Xenograft tumors of ETK cells in vivo accumulated 2.5-fold more 8-[3H]GCV per gram of tissue and showed greater fluorescence from EGFP than control DU145 cells, demonstrating that the reporter gene functioned in vivo. These data extend previous reports by showing that a human promoter can be detected in vitro and in vivo with a dual-function reporter exploiting optical and radiotracer techniques.


Assuntos
Proteínas Luminescentes/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Timidina Quinase/metabolismo , Sequência de Bases , Primers do DNA , Citometria de Fluxo/métodos , Genes Reporter , Proteínas de Fluorescência Verde , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Humanos , Cinética , Proteínas Luminescentes/genética , Masculino , Microscopia de Fluorescência/métodos , Reação em Cadeia da Polimerase , Neoplasias da Próstata , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina Quinase/genética , Transfecção , Células Tumorais Cultivadas
8.
Mol Imaging ; 1(1): 24-35, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12920858

RESUMO

Multidrug resistance (MDR) mediated by overexpression of MDR1 P-glycoprotein (Pgp) is one of the best characterized barriers to chemotherapy in cancer patients. Furthermore, the protective function of Pgp-mediated efflux of xenobiotics in various organs has a profound effect on the bioavailability of drugs in general. Thus, there is an expanding requirement to noninvasively interrogate Pgp transport activity in vivo. We herein report the Pgp recognition properties of a novel 99mTc(I)-tricarbonyl complex, [99mTc(CO)3(MIBI)3]+ (Tc-CO-MIBI). Tc-CO-MIBI showed 60-fold higher accumulation in drug-sensitive KB 3-1 cells compared to colchicine-selected drug-resistant KB 8-5 cells. In KB 8-5 cells, tracer enhancement was observed with the potent MDR modulator LY335979 (EC50 = 62 nM). Similar behavior was observed using drug-sensitive MCF-7 breast adenocarcinoma cells and MCF-7/MDR1 stable transfectants, confirming that Tc-CO-MIBI is specifically excluded by overexpression of MDR1 Pgp. By comparison, net accumulation in control H69 lung tumor cells was 9-fold higher than in MDR-associated protein (MRP1)-expressing H69AR cells, indicating only modest transport by MRP1. Biodistribution analysis following tail vein injection of Tc-CO-MIBI showed delayed liver clearance as well as enhanced brain uptake and retention in mdr1a/1b(-/-) gene deleted mice versus wild-type mice, directly demonstrating that Tc-CO-MIBI is a functional probe of Pgp transport activity in vivo.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Encéfalo/metabolismo , Compostos de Organotecnécio , Tecnécio Tc 99m Sestamibi , Adenocarcinoma , Animais , Transporte Biológico , Neoplasias da Mama , Humanos , Células KB , Cinética , Neoplasias Pulmonares , Camundongos , Camundongos Knockout , Modelos Moleculares , Conformação Molecular , Compostos Radiofarmacêuticos , Proteínas Recombinantes/metabolismo , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA