Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
FEBS J ; 286(8): 1543-1560, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30715798

RESUMO

Double-stranded RNA (dsRNA) is a potent proinflammatory signature of viral infection and is sensed primarily by RIG-I-like receptors (RLRs). Oligomerization of RLRs following binding to cytosolic dsRNA activates and nucleates self-assembly of the mitochondrial antiviral-signaling protein (MAVS). In the current signaling model, the caspase recruitment domains of MAVS form helical fibrils that self-propagate like prions to promote signaling complex assembly. However, there is no conclusive evidence that MAVS forms fibrils in cells or with the transmembrane anchor present. We show here with super-resolution light microscopy that MAVS activation by dsRNA induces mitochondrial membrane remodeling. Quantitative image analysis at imaging resolutions as high as 32 nm shows that in the cellular context, MAVS signaling complexes and the fibrils within them are smaller than 80 nm. The transmembrane domain of MAVS is required for its membrane remodeling, interferon signaling, and proapoptotic activities. We conclude that membrane tethering of MAVS restrains its polymerization and contributes to mitochondrial remodeling and apoptosis upon dsRNA sensing.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Interferon beta/metabolismo , Membranas Mitocondriais/metabolismo , Células 3T3/virologia , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Morte Celular/fisiologia , Citosol/fisiologia , Fibroblastos/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Knockout , Microscopia/métodos , Membranas Mitocondriais/virologia , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Domínios Proteicos , RNA de Cadeia Dupla/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Análise de Célula Única/métodos , Febre do Nilo Ocidental/metabolismo
2.
Mol Biol Cell ; 18(4): 1253-60, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17267685

RESUMO

Protein disulfide isomerase (PDI)-like proteins act as oxido-reductases and chaperones in the endoplasmic reticulum (ER). How oligomerization of the PDI-like proteins control these activities is unknown. Here we show that dimerization of ERp29, a PDI-like protein, regulates its protein unfolding and escort activities. We have demonstrated previously that ERp29 induces the local unfolding of polyomavirus in the ER, a step required for viral infection. We now find that, in contrast to wild-type ERp29, a mutant ERp29 (D42A) that dimerizes inefficiently is unable to unfold polyomavirus or stimulate infection. A compensatory mutation that partially restores dimerization to the mutant ERp29 (G37D/D42A) rescues ERp29 activity. These results indicate that dimerization of ERp29 is crucial for its protein unfolding function. ERp29 was also suggested to act as an escort factor by binding to the secretory protein thyroglobulin (Tg) in the ER, thereby facilitating its secretion. We show that this escort function likewise depends on ERp29 dimerization. Thus our data demonstrate that dimerization of a PDI-like protein acts to regulate its diverse ER activities.


Assuntos
Proteínas de Choque Térmico/metabolismo , Células 3T3/virologia , Animais , Células Cultivadas , Dimerização , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/genética , Camundongos , Mutação , Polyomavirus/patogenicidade , Infecções por Polyomavirus/metabolismo , Estrutura Terciária de Proteína , Ratos , Tireoglobulina/metabolismo
3.
Glycoconj J ; 23(3-4): 189-97, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16691502

RESUMO

Our previous studies show that the depletion of cholesterol or sphingolipids (raft-associated lipids) from receptor-bearing adherent cell lines blocks HIV-1 entry and HIV-1 Env-mediated membrane fusion. Here we have evaluated the mechanism(s) by which these lipids contribute to the HIV-1 Env-mediated membrane fusion. We report the following: (1) GSL depletion from a suspension T lymphocyte cell line (Sup-T1) reduced subsequent fusion with HIV-1IIIB-expressing cells by 70%. (2) Cholesterol depletion from NIH3T3 cells bearing HIV-1 receptors (NIH3T3CD4R5/NIH3T3CD4X4) did not impair subsequent fusion with HeLa cells expressing the corresponding HIV-1 Envs. In contrast GSL depletion from these targets reduced fusion by 50% suggesting that GSL facilitate fusion in different ways. (3) GSL-deficient GM95 cells bearing high receptors fused with HIV-1 Env-expressing cells at 37 degrees C with kinetics similar to that of GSL + NIH3T3 targets. Based on these observations, we propose that the plasma membrane cholesterol is required to maintain the integrity of receptor pools whereas GSLs are involved in stabilizing the coupling of inter-receptor pools.


Assuntos
Colesterol/metabolismo , HIV-1/fisiologia , HIV-1/patogenicidade , Fusão de Membrana/fisiologia , Esfingolipídeos/metabolismo , Células 3T3/virologia , Animais , Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Glicoesfingolipídeos/metabolismo , Humanos , Cinética , Fusão de Membrana/efeitos dos fármacos , Camundongos , Morfolinas/farmacologia , Receptores de HIV/efeitos dos fármacos , Receptores de HIV/metabolismo , Esfingolipídeos/farmacologia
4.
Biochem J ; 381(Pt 2): 437-46, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15025565

RESUMO

Early events play a decisive role in virus multiplication. We have shown previously that activation of MAPK/ERK1/2 (mitogen-activated protein kinase/extracellular-signal-regulated kinase 1/2) and protein kinase A are pivotal for vaccinia virus (VV) multiplication [de Magalhães, Andrade, Silva, Sousa, Ropert, Ferreira, Kroon, Gazzinelli and Bonjardim (2001) J. Biol. Chem. 276, 38353-38360]. In the present study, we show that VV infection provoked a sustained activation of both ERK1/2 and RSK2 (ribosomal S6 kinase 2). Our results also provide evidence that this pattern of kinase activation depends on virus multiplication and ongoing protein synthesis and is maintained independently of virus DNA synthesis. It is noteworthy that the VGF (VV growth factor), although involved, is not essential for prolonged ERK1/2 activation. Furthermore, our findings suggest that the VV-stimulated ERK1/2 activation also seems to require actin dynamics, microtubule polymerization and tyrosine kinase phosphorylation. The VV-stimulated pathway MEK/ERK1/2/RSK2 (where MEK stands for MAPK/ERK kinase) leads to phosphorylation of the ternary complex factor Elk-1 and expression of the early growth response (egr-1) gene, which kinetically paralleled the kinase activation. The recruitment of this pathway is biologically relevant, since its disruption caused a profound effect on viral thymidine kinase gene expression, viral DNA replication and VV multiplication. This pattern of sustained kinase activation after VV infection is unique. In addition, by connecting upstream signals generated at the cytoskeleton and by tyrosine kinase, the MEK/ERK1/2/RSK2 cascade seems to play a decisive role not only at early stages of the infection, i.e. post-penetration, but is also crucial to define the fate of virus progeny.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/fisiologia , Vaccinia virus/enzimologia , Replicação Viral/fisiologia , Células 3T3/enzimologia , Células 3T3/virologia , Actinas/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Replicação do DNA/genética , DNA Viral/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Viral da Expressão Gênica/genética , Genes Precoces/genética , Proteínas Imediatamente Precoces/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Peptídeos/fisiologia , Fosforilação , Biossíntese de Proteínas/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Células Vero/virologia , Proteínas Elk-1 do Domínio ets
5.
Blood ; 102(13): 4535-40, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12920032

RESUMO

The ability of viral or mutated cellular oncogenes to initiate neoplastic events and their poor immunogenicity have considerably undermined their potential use as immunotherapeutic tools for the treatment of human cancers. Using an Epstein-Barr virus-encoded oncogene, latent membrane protein 1 (LMP1), as a model, we report a novel strategy that both deactivates cellular signaling pathways associated with the oncogenic phenotype and reverses poor immunogenicity. We show that cotranslational ubiquitination combined with N-end rule targeting of LMP1 enhanced the intracellular degradation of LMP1 and total blockade of LMP1-mediated nuclear factor-kappaB (NF-kappaB) and signal transducer and activator of transcription (STAT) activation in human cells. In addition, although murine cells expressing LMP1 were uniformly tumorigenic, this oncogenicity was completely abrogated by covalent linkage of LMP1 with ubiquitin, while an enhanced CD8+ T cell response to a model epitope fused to the C-terminus of LMP1 was observed following immunization with ubiquitinated LMP1. These observations suggest that proteasomal targeting of tumor-associated oncogenes could be exploited therapeutically by either gene therapy or vaccination.


Assuntos
Cisteína Endopeptidases/metabolismo , Genes Virais , Herpesvirus Humano 4/imunologia , Complexos Multienzimáticos/metabolismo , Oncogenes , Processamento de Proteína Pós-Traducional , Proteínas da Matriz Viral/metabolismo , Células 3T3/patologia , Células 3T3/transplante , Células 3T3/virologia , Animais , Linfócitos T CD8-Positivos/imunologia , Transformação Celular Viral , Epitopos/imunologia , Antígenos H-2/imunologia , Humanos , Masculino , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Neoplasias Experimentais/etiologia , Fenótipo , Complexo de Endopeptidases do Proteassoma , Proteínas Recombinantes de Fusão/imunologia , Transcrição Gênica , Transfecção , Ubiquitina/metabolismo , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/imunologia
6.
J Gen Virol ; 84(Pt 7): 1677-1686, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12810861

RESUMO

Cultivation of retrovirus packaging cells at 32 degrees C represents a common procedure to achieve high titres in mouse retrovirus production. Gene expression profiling of mouse NIH 3T3 cells producing amphotropic mouse leukaemia virus 4070A revealed that 10 % of the 1176 cellular genes investigated were regulated by temperature shift (37/32 degrees C), while 5 % were affected by retrovirus infection. Strikingly, retrovirus production at 32 degrees C activated the cholesterol biosynthesis/transport pathway and caused an increase in plasma membrane cholesterol levels. Furthermore, these conditions resulted in transcriptional activation of smoothened (smo), patched (ptc) and gli-1; Smo, Ptc and Gli-1, as well as cholesterol, are components of the Sonic hedgehog (Shh) signalling pathway, which directs pattern formation, diversification and tumourigenesis in mammalian cells. These findings suggest a link between cultivation at 32 degrees C, production of MLV-A and the Shh signalling pathway.


Assuntos
Células 3T3/virologia , Vírus da Leucemia Murina/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas/metabolismo , Temperatura , Montagem de Vírus , Células 3T3/fisiologia , Animais , Membrana Celular/química , Membrana Celular/metabolismo , Colesterol/análise , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas Hedgehog , Vírus da Leucemia Murina/patogenicidade , Camundongos , Proteínas/genética , Transdução de Sinais , Transativadores/metabolismo
7.
Int J Cancer ; 105(5): 644-53, 2003 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-12740913

RESUMO

Epstein-Barr virus (EBV) carrying lymphoblastoid cells of normal origin express the full program of all 9 virus-encoded, growth transformation associated proteins. They have an intact p53 pathway as a rule. This raises the question of whether any of the viral proteins impair the pathway functionally. Using a yeast 2-hybrid system, we have shown that EBNA-5 but not the other EBNAs interacts with the p14ARF protein, a regulator of the p53 pathway. The interaction was confirmed in vitro using a GST pull-down assay. Moreover, expression of EBNA-5 increased the survival of p14ARF-transfected cells. EBV infection of resting B cells induced the expression of p14ARF mRNA without increased level of the protein. A fraction of the p14ARF localized to the nucleoli but the bulk of the protein accumulated in nuclear but extranucleolar inclusions. Formation of the extranucleolar inclusions led to complete relocalization of EBNA-5 from nucleoplasm to these structures. The inclusions also contained p53 and HDM2, and were surrounded by PML bodies and proteasomes, which suggests that these inclusions could be targets for proteasome dependent protein degradation.


Assuntos
Linfócitos B/virologia , Núcleo Celular/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/fisiologia , Herpesvirus Humano 4/fisiologia , Proteínas Nucleares , Proteína Supressora de Tumor p14ARF/metabolismo , Células 3T3/metabolismo , Células 3T3/virologia , Transporte Ativo do Núcleo Celular , Adenocarcinoma/patologia , Animais , Linfócitos B/metabolismo , Linfócitos B/ultraestrutura , Neoplasias Ósseas/patologia , Nucléolo Celular/metabolismo , Sobrevivência Celular , Transformação Celular Viral/fisiologia , Neoplasias do Colo/patologia , Cisteína Endopeptidases/metabolismo , Regulação Viral da Expressão Gênica , Células HeLa/metabolismo , Células HeLa/virologia , Humanos , Camundongos , Complexos Multienzimáticos/metabolismo , Osteossarcoma/patologia , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/virologia , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/metabolismo , Técnicas do Sistema de Duplo-Híbrido
8.
J Virol ; 77(5): 2850-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12584308

RESUMO

The ovine betaretroviruses jaagsiekte sheep retrovirus (JSRV) and enzootic nasal tumor virus (ENTV) cause contagious cancers in the lungs and upper airways of sheep and goats. Oncogenic transformation assays using mouse and rat fibroblasts have localized the transforming activity to the Env proteins encoded by these viruses, which require the putative lung and breast cancer tumor suppressor hyaluronidase 2 (Hyal2) to promote virus entry into cells. These results suggested the hypothesis that the JSRV and ENTV Env proteins cause cancer by inhibiting the tumor suppressor activity of Hyal2. Consistent with this hypothesis, we show that human Hyal2 and other Hyal2 orthologs that can promote virus entry, including rat Hyal2, can suppress transformation by the Env proteins of JSRV and ENTV. Furthermore, we provide direct evidence for binding of the surface (SU) region of JSRV Env to human and rat Hyal2. However, mouse Hyal2 did not mediate entry of virions bearing JSRV or ENTV Env proteins, bound JSRV SU poorly if at all, and did not suppress transformation by the JSRV or ENTV Env proteins, indicating that mouse Hyal2 plays no role in transformation of mouse fibroblasts and that the Env proteins can transform at least some cells by a Hyal2-independent mechanism. Expression of human Hyal2 in mouse cells expressing JSRV Env caused a marked reduction in Env protein levels, indicating that human Hyal2 suppresses Env-mediated transformation in mouse cells by increasing Env degradation rather than by exerting a more general Env-independent tumor suppressor activity.


Assuntos
Betaretrovirus/fisiologia , Transformação Celular Viral , Produtos do Gene env/metabolismo , Hialuronoglucosaminidase/metabolismo , Células 3T3/virologia , Animais , Betaretrovirus/genética , Betaretrovirus/metabolismo , Linhagem Celular , Clonagem Molecular , Fibroblastos/virologia , Humanos , Hialuronoglucosaminidase/genética , Retrovirus Jaagsiekte de Ovinos/genética , Retrovirus Jaagsiekte de Ovinos/metabolismo , Retrovirus Jaagsiekte de Ovinos/fisiologia , Camundongos , Dados de Sequência Molecular , Ratos , Receptores Virais/metabolismo , Análise de Sequência de DNA
9.
J Virol ; 77(4): 2615-22, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12552000

RESUMO

The murine polyomavirus (Py) enters mouse fibroblasts and kidney epithelial cells via an endocytic pathway that is caveola-independent (as well as clathrin-independent). In contrast, uptake of simian virus 40 into the same cells is dependent on caveola. Following the initial uptake of Py, both microtubules and microfilaments play roles in trafficking of the virus to the nucleus. Colcemid, which disrupts microtubules, inhibits the ability of Py to reach the nucleus and replicate. Paclitaxel, which stabilizes microtubules and prevents microtubule turnover, has no effect, indicating that intact but not dynamic microtubules are required for Py infectivity. Compounds that disrupt actin filaments enhance Py uptake while stabilization of actin filaments impedes Py infection. Virus particles are seen in association with actin in cells treated with microfilament-disrupting or filament-stabilizing agents at levels comparable to those in untreated cells, suggesting that a dynamic state of the microfilament system is important for Py infectivity.


Assuntos
Polyomavirus/fisiologia , Polyomavirus/patogenicidade , Vírus 40 dos Símios/fisiologia , Vírus 40 dos Símios/patogenicidade , Células 3T3/virologia , Actinas/metabolismo , Animais , Cavéolas/efeitos dos fármacos , Cavéolas/virologia , Células Cultivadas , Citoesqueleto/metabolismo , Demecolcina/farmacologia , Células Epiteliais/virologia , Rim/citologia , Rim/virologia , Camundongos , Microtúbulos , Nistatina/farmacologia , Paclitaxel/farmacologia
10.
Hum Gene Ther ; 13(14): 1783-90, 2002 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-12396630

RESUMO

Guidelines for testing gene therapy products for ecotropic replication-competent retrovirus (Eco-RCR) have not been delineated as they have for amphotropic viruses. To evaluate biologic assays that can detect these viruses, we compared an S(+)/L(-) assay and a marker rescue assay designed specifically for Eco-RCR detection. Moloney murine leukemia virus (Mo-MuLV) obtained from the American Type Culture Collection was used as the positive control. For marker rescue, NIH 3T3 cells were transduced with a retroviral vector expressing the neomycin phosphotransferase gene (3T3/Neo). Inoculation and passage of test material in 3T3/Neo cells for 3 weeks (amplification) and subsequent testing in the S(+)/L(-) assay or the marker rescue assay increased the level of sensitivity for virus detection greater than 10-fold compared with direct inoculation of D56 S(+)/L(-) cells. When serial dilutions of Mo-MuLV stock were evaluated, six of six cultures had detectable virus by the S(+)/L(-) and marker rescue assays at dilutions of 10(-5) and 10(-6). At the 10(-7) dilution, five of six assays had detectable virus in both assays. The ability to detect virus-infected cells was also evaluated in a modification that substituted cells for supernatant. Fifteen 3T3/Neo cultures inoculated with 10(6) 293 cells containing 100 or 10 Mo-MuLV/3T3 cells were all positive by marker rescue. For dilution with 1 virus-infected cell per 10(6) 293 cells, 10 of 15 cultures were positive. At the 0.1-cell dilution only 2 of 15 cultures were positive. If we hope to detect one infected cell in a test article, the probability of detecting virus if the assay is performed in triplicate is 96.3%. In summary, after 3 weeks of amplification the S(+)/L(-) and marker rescue assays can detect virus with similar sensitivities. We prefer the marker rescue assay because of the more reliable growth features of NIH 3T3 cells compared with the D56 cell line. For laboratories analyzing clinical materials, this report may prove useful in establishing detection assays for Eco-RCR.


Assuntos
Bioensaio , Terapia Genética/normas , Vetores Genéticos/fisiologia , Canamicina Quinase/análise , Retroviridae/isolamento & purificação , Vírus Formadores de Foco no Baço/isolamento & purificação , Replicação Viral , Células 3T3/virologia , Animais , Linhagem Celular/virologia , Vírus Defeituosos/fisiologia , Genes Reporter , Marcadores Genéticos , Humanos , Canamicina Quinase/genética , Rim , Camundongos , Vírus da Leucemia Murina de Moloney/fisiologia , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Retroviridae/fisiologia , Segurança , Vírus do Sarcoma Murino/fisiologia , Sensibilidade e Especificidade , Vírus Formadores de Foco no Baço/fisiologia , Transdução Genética
11.
Oncogene ; 21(36): 5574-81, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12165856

RESUMO

The late region of the human neurotropic JC virus encodes a 71 amino acid protein, named Agnoprotein, whose biological function remains elusive. Here we demonstrate that in the absence of other viral proteins, expression of Agnoprotein can inhibit cell growth by deregulating cell progression through the cell cycle stages. Cells with constitutive expression of Agnoprotein were largely accumulated at the G2/M stage and that decline in the activity of cyclins A and B is observed in these cells. Agnoprotein showed the ability to augment p21 promoter activity in transient transfection assay and a noticeable increase in the level of p21 is detected in cells continuously expressing Agnoprotein. Results from binding studies revealed the interaction of Agnoprotein with p53 through the N-terminal of the Agnoprotein spanning residues 1-36. Co-expression of p53 and Agnoprotein further stimulated transcription of the p21 promoter. Thus, the interaction of p53 and Agnoprotein can lead to a higher level of p21 expression and suppression of cell cycle progression during the cell cycle.


Assuntos
Vírus JC/fisiologia , Proteínas Virais/fisiologia , Células 3T3/metabolismo , Células 3T3/virologia , Animais , Proteínas de Bactérias/metabolismo , Western Blotting , Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Primers do DNA/química , Inibidores Enzimáticos/metabolismo , Citometria de Fluxo , Regulação Viral da Expressão Gênica , Glutationa Transferase/metabolismo , Humanos , Proteínas Luminescentes/metabolismo , Camundongos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/fisiologia , Proteínas Quinases/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Virais Reguladoras e Acessórias
12.
Ann Biomed Eng ; 30(5): 731-42, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12108847

RESUMO

The overall dynamics of the retrovirus-cell encounter under a static retroviral transduction system can be described in terms of the process of uptake (adsorption/ internalization), decay, and diffusion. In this study, a mathematical model illustrating these processes was derived assuming a semi-infinite domain and solved analytically using the Laplace transform. The closed-form solutions for retroviral concentrations and time course profile of transduced cell colonies are presented to clarify the contributions of the processes involved in the retroviral transduction system. To manifest the usefulness of the closed-form solutions, the neomycin-resistant gene encoding retroviruses produced by two different packaging cells (human 293 cells and murine GP+ E86/LNCX cells) were employed to transduce NIH 3T3 cells, which formed neomycin-resistant colonies after G418 selection. The experimental results were curve fitted with the model-derived analytical solutions to quantitatively determine transduction rate constant k and initial concentration of infectious retrovirus C0. Our study showed that the vesicular stomatitis virus G protein pseudotyped retrovirus produced from 293 packaging cells exhibited much higher transduction rate (k = 0.0480 cm/h) than the ecotropic retrovirus (k = 0.0102 cm/h) produced from GP + E86/LNCX cells. The fitted values of C0 are approximately two orders of magnitude higher than the experimentally estimated titers for both retroviruses.


Assuntos
Células 3T3/fisiologia , Células 3T3/virologia , Vírus da Leucemia Murina/genética , Modelos Genéticos , Retroviridae/fisiologia , Transdução Genética , Animais , Adesão Celular , Linhagem Celular , Movimento Celular/fisiologia , Simulação por Computador , Vetores Genéticos/genética , Meia-Vida , Camundongos , Retroviridae/genética , Integração Viral
13.
Genome Res ; 12(4): 595-601, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11932243

RESUMO

The nucleotide sequence of chicken Mx cDNA was reported earlier using the White Leghorn breed in Germany, but it showed no enhanced resistance to viruses. In this study, the nucleotide sequences of chicken Mx cDNA were determined in many breeds. A total of 25 nucleotide substitutions, of which 14 were deduced to cause amino acid exchanges, were detected, suggesting that the chicken Mx gene is very polymorphic. Transfected cell clones expressing chicken Mx mRNA were established after the Mx cDNA was constructed with an expression vector and introduced into mouse 3T3 cells, and the Mx genes from some breeds were demonstrated to confer positive antiviral responses to influenza virus and vesicular stomatitis virus. On the basis of the comparison among the antiviral activities associated with many Mx variations, a specific amino acid substitution at position 631 (Ser to Asn) was considered to determine the antivirally positive or negative Mx gene. Thus, a single amino acid substitution influences the antiviral activity of Mx in domesticated chickens.


Assuntos
Antivirais/genética , Galinhas/genética , Proteínas de Ligação ao GTP , Polimorfismo Genético/genética , Proteínas/genética , Células 3T3/virologia , Substituição de Aminoácidos/genética , Animais , Asparagina/genética , Células Cultivadas , Embrião de Galinha/virologia , Regulação da Expressão Gênica/fisiologia , Variação Genética , Vetores Genéticos/biossíntese , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Vírus da Influenza A/crescimento & desenvolvimento , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Proteínas de Resistência a Myxovirus , Filogenia , Biossíntese de Proteínas , Proteínas/fisiologia , Serina/genética , Transfecção , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Cultura de Vírus
14.
APMIS ; 110(12): 899-912, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12645669

RESUMO

FIS-2, a less oncogenic, immunosuppressive variant of the Friend murine leukaemia virus (F-MuLV), was used to explore whether the differences in biological features were related to early virus dissemination rates or sites of replication. We found that erythroblasts were the primary target cells for both F-MuLV and FIS-2, while B- and T-cells were infected later in the infection. Although FIS-2 replicated to similar titres as F-MuLV, we observed a delay in peak viraemia titre and in the number of virus-positive cells in bone marrow and spleen. Studies including the chimeric viruses RE3 (FIS-2LTR with a F-MuLV background) and RE4 (F-MuLV LTR with a FIS-2 background) indicated that the delay in dissemination was due to mutations in FIS-2 LTR. The kinetics for early virus replication correlated with previously reported mean latency time for virus-induced erythroleukaemia in mice inoculated as newborns and with the onset of immunosuppression in adult mice. In addition, F-MuLV-induced late immunosuppression coincided with signs of erythroleukaemia and persistent viraemia. FIS-2 induced a more moderate late immunosuppression without persistent viraemia or signs of erythroleukaemia. Overall, our findings indicated that early viral replication is a prognostic factor in murine retrovirus-induced pathogenesis.


Assuntos
Vírus da Leucemia Murina de Friend/patogenicidade , Vírus Auxiliares/patogenicidade , Síndromes de Imunodeficiência/virologia , Leucemia Eritroblástica Aguda/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Viremia/virologia , Células 3T3/virologia , Fatores Etários , Animais , Animais Recém-Nascidos , Linfócitos B/virologia , Eritroblastos/virologia , Feminino , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/fisiologia , Vírus Auxiliares/genética , Vírus Auxiliares/fisiologia , Camundongos , RNA Viral/sangue , Baço/virologia , Linfócitos T/virologia , Sequências Repetidas Terminais/genética , Virulência , Replicação Viral
15.
J Antimicrob Chemother ; 48(6): 757-67, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11733458

RESUMO

For two decades it has been impossible to develop drugs with novel mechanisms of action against herpesviruses, and treatment has been confined largely to the use of inhibitors of viral DNA polymerase. As a representative of a novel inhibitory approach, the non-nucleosidic BAY 38-4766 was identified as a highly selective inhibitor of human cytomegalovirus (HCMV). The compound selectively inhibits not only HCMV strains, including ganciclovir-resistant, ganciclovir/foscarnet and ganciclovir/cidofovir double-resistant clinical isolates, but also a number of monkey and rodent cytomegaloviruses. In a murine cytomegalovirus (MCMV) pathogenicity model in mice, antiviral efficacy and excellent tolerability were demonstrated. BAY 38-4766-resistant HCMV and MCMV strains are not cross-resistant to the nucleoside analogues ganciclovir and cidofovir or the pyrophosphate analogue foscarnet, indicating a different mode of action. Mechanistic studies demonstrated that the high selectivity of this drug class is most likely due to the inhibition of a late stage of the viral replication cycle. Sequence analyses of resistant HCMV and MCMV strains revealed mutations in UL89 and UL104, proteins known to be involved in viral DNA cleavage and packaging. Consequently, the drug is highly specific for the viral as opposed to cellular functions, since UL89 is related to a bacteriophage terminase and no human equivalent exists. In addition, because some of the genes of the viral DNA cleavage and packaging complex are highly conserved among herpesviruses, development of broad-spectrum agents covering additional human herpesviruses might be possible using this approach.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Naftalenossulfonatos/farmacologia , Células 3T3/efeitos dos fármacos , Células 3T3/virologia , Animais , Linhagem Celular , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/virologia , DNA Viral/genética , DNA Viral/isolamento & purificação , Relação Dose-Resposta a Droga , Farmacorresistência Viral/genética , Humanos , Masculino , Camundongos , Camundongos SCID , Testes de Sensibilidade Microbiana/métodos
16.
AIDS Res Hum Retroviruses ; 17(16): 1489-500, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11709093

RESUMO

Cell motility is likely to play a pivotal role in HIV infection by promoting the dissemination of infected cells. On the basis of observations indicating an interaction between HIV-1 Gag and target cell filamentous actin, we hypothesized that these interactions would promote cell motility of HIV-infected cells. Indeed, we have found that HIV-1 infection enhances the chemotactic response of macrophages. To specifically investigate the significance of the interactions between Gag and cellular actin, we transfected NIH 3T3 fibroblasts and HeLa cells with a construct that permits the expression of HIV-1 Gag in the absence of any other viral protein. Fractionation experiments showed that Gag was present in cytoskeletal fraction containing long actin filaments and in a high-speed postcytoskeletal fraction with short actin filaments. We have also localized HIV-1 Gag to the lamellipodia of chemoattractant-stimulated cells. Significantly, the motility of Gag-expressing cells was enhanced in chemotaxis assays. In vitro mutagenesis experiments showed that HIV-1 Gag binds filamentous actin through the nucleocapsid domain (NC). An NC-green fluorescent protein fusion had the same cellular distribution as the complete protein, and its expression increased cell motility. These data suggest that interactions between HIV-1 Gag and actin in infected cells enhance cell motility. Ultimately this enhanced motility of infected cells could promote the dissemination of virus into the brain and other tissues.


Assuntos
Quimiotaxia/fisiologia , Citoesqueleto/metabolismo , Produtos do Gene gag/metabolismo , HIV-1/patogenicidade , Macrófagos/fisiologia , Nucleocapsídeo/metabolismo , Células 3T3/fisiologia , Células 3T3/virologia , Actinas/metabolismo , Animais , Produtos do Gene gag/genética , HIV-1/fisiologia , Células HeLa/fisiologia , Células HeLa/virologia , Humanos , Macrófagos/virologia , Camundongos , Monócitos/fisiologia , Monócitos/virologia , Transfecção
17.
Nat Cell Biol ; 3(8): 745-50, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11483960

RESUMO

The importance of herpes simplex viruses (HSV) as human pathogens and the emerging prospect of using mutant derivatives of HSV-1 as potential anti-cancer therapeutics have necessitated a thorough investigation into the molecular basis of host-cell permissiveness to HSV. Here we show that NIH-3T3 cells transformed with the oncogenes v-erbB, activated sos or activated ras become significantly more permissive to HSV-1. Inhibitors of the Ras signalling pathway, such as farnesyl transferase inhibitor 1 and PD98059, effectively suppressed HSV-1 infection of ras-transformed cells. Enhanced permissiveness of the transformed cells was linked to the inhibition of virus-induced activation (phosphorylation) of the double-stranded RNA-activated protein kinase (PKR), thereby allowing viral transcripts to be translated in these cells. An HSV-1-derived oncolytic mutant, R3616, was also found to infect preferentially both transformed cells and PKR-/- (but not PKR+/+) mouse embryo fibroblasts. These observations suggest that HSV-1 specifically targets cells with an activated Ras signalling pathway, and have important ramifications in the use of engineered HSV in cancer therapy, the development of strategies against HSV infections, and the controversial role of HSV in human cancers.


Assuntos
Transformação Celular Viral/genética , Herpesvirus Humano 1/patogenicidade , Interações Hospedeiro-Parasita/genética , Oncogenes/fisiologia , Transdução de Sinais/genética , Proteínas ras/metabolismo , Células 3T3/citologia , Células 3T3/metabolismo , Células 3T3/virologia , Alquil e Aril Transferases/antagonistas & inibidores , Animais , Linhagem Celular Transformada/citologia , Linhagem Celular Transformada/metabolismo , Linhagem Celular Transformada/virologia , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase , Flavonoides/farmacologia , Genes erbB-1/genética , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , MAP Quinase Quinase 1 , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação/fisiologia , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , Proteína Son Of Sevenless de Drosófila/genética , Proteínas Virais/biossíntese , Proteínas Virais/genética , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo , Proteínas ras/genética
18.
Arch Virol ; 146(5): 953-61, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11448032

RESUMO

We previously obtained a chimeric Friend murine leukemia virus (FMLV) envelope protein (Env) in which the whole receptor-binding domain (RBD) was replaced with a surface domain of human CD4. Here, we examined if the postbinding fusion function of the CD4-Env chimera still remains to be intact. While a pseudotype MLV bearing CD4-Env showed no infectivity, NIH 3T3 cells could be infected with a pseudotype MLV bearing both CD4-Env and a mutant FMLV Env defective in postbinding fusion function. The pseudotype MLV showed no infectivity on HeLa cells but on the FMLV receptor (mCAT1)-expressing HeLa cells. In NIH 3T3 cells, the R-peptide-deleted CD4-Env could not induce syncytia by itself but did so in co-operation with the fusion-deficient Env. Syncytia induced by the coexpression were not observed in HeLa cells but in the mCAT1-expressing HeLa cells. These results indicate that the CD4-Env could contribute postbinding fusion function in the membrane fusion process triggered by FMLV RBD-mCAT1 interaction.


Assuntos
Antígenos CD4/genética , Vírus da Leucemia Murina de Friend/fisiologia , Produtos do Gene env/fisiologia , Fusão de Membrana/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Células 3T3/virologia , Animais , Sítios de Ligação , Efeito Citopatogênico Viral/genética , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/patogenicidade , Genes env , Células Gigantes/virologia , Células HeLa/virologia , Humanos , Fusão de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Mutação Puntual , Estrutura Terciária de Proteína , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/genética , Virulência , Cultura de Vírus
19.
Jpn J Cancer Res ; 92(4): 410-6, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11346463

RESUMO

Human T-cell leukemia virus type 1 (HTLV-1) is the etiologic agent for adult T-cell leukemia and HTLV-1-associated myelopathy / tropical spastic paraparesis. Recently we infected newborn mice by inoculating HTLV-1-producing human cells, and found that T-cells, B-cells and granulocytes were infected in vivo. To understand the mechanism of viral-cell interaction and the pathogenesis of HTLV-1 using the mouse model, it is important to clarify the cellular tropism using a cell-free HTLV-1 transmission system. We employed a highly transmissible cell-free HTLV-1 produced by a feline kidney cell line, c77, and studied the susceptibility of 9 kinds of mouse cell lines, EL4, RLm1, CTLL-2, J774.1, DA-1, Ba / F3, WEHI-3, NIH3T3 and B1, and two kinds of human cell lines, Molt-4 and Hut78. HTLV-1 proviral sequence was found by PCR in all 9 mouse cell lines as well as in 2 human cell lines and viral entry was blocked with sera from an HTLV-1 carrier and an adult T-cell leukemia patient. Unexpectedly, mouse cell lines EL4 and RLm1 and human cell lines Molt-4 and Hut78 showed similar efficiency for viral entry. These results suggest a wide distribution of HTLV-1 receptor in mouse cells.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Células 3T3/virologia , Animais , Anticorpos Antivirais/imunologia , Portador Sadio/sangue , Portador Sadio/imunologia , Portador Sadio/virologia , Gatos , Linhagem Celular , Sistema Livre de Células , DNA Viral/análise , Produtos do Gene gag/biossíntese , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Humanos , Leucemia-Linfoma de Células T do Adulto/sangue , Leucemia-Linfoma de Células T do Adulto/imunologia , Leucemia-Linfoma de Células T do Adulto/virologia , Camundongos , Reação em Cadeia da Polimerase , Proteínas Oncogênicas de Retroviridae/biossíntese , Linfócitos T/virologia , Vírion/patogenicidade , Produtos do Gene gag do Vírus da Imunodeficiência Humana
20.
J Tongji Med Univ ; 21(3): 215-8, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12539580

RESUMO

The bioactivities of culture supernatants from retroviral packaging cells carrying the mouse Fas ligand (mFasL) gene was investigated. FasLcDNA was cloned into PLXIN with an internal ribosome entry site to link two cistrons through gene recombination technology, PLXIN and the recombinant vector PLFIN were separately transfected into PA317 retrovirus packing cell line by lipofectamine 2000, and the resistant clones were selected with G418 selective medium. The integration of genome DNA was assayed by genomic DNA PCR. NIH3T3 cells were transduced by the culture supernatants from PA317 carrying the mFasLcDNA gene, and were selected with G418 selective medium, so as to select the PLFIN-PA317 clone capable of producing higher titer of supernatants. The levels of mFasL protein on NIH3T3 cells membrane were assayed by flow cytometry (FCM). The biological activity of mFasL on NIH3T3 cells membrane was investigated by the inducing apoptosis of Fas+ Yac-1 cells co-cultured with NIH3T3 cells expressing Fas ligand. To explore the direct mFasL cytotoxicity of culture supernatants from retroviral packaging cells carrying the mFasL gene, the culture supernatants from PLFIN-PA317 and PLXIN-PA317 were separately co-cultured with Yac-1 cells in parallel. The recombinant PLFIN was successfully constructed. The highest titer of supernatants from twelve resistant clones was 8.5 x 10(5) colony-forming-unit (CFU)/ml. The NIH3T3 cells transfected by above supernatants had a higher level of mFasL (53.81 +/- 6.9%), and significantly induced the apoptosis of Fas+ Yac-1 cells (56.78 +/- 4.5%), as both were cocultured for 5 h at 1:1 ratio, whereas it is 7.08 +/- 3.4% in control group (P < 0.01). Supernatant from PLFIN-PA317 could also directly induce the apoptosis of Yac-1 within 5 h of incubation. Thus, the culture supernatants from PLFIN-PA317 possessed both infectivity and cytotoxicity of mFasL.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Glicoproteínas de Membrana/genética , Retroviridae/genética , Células 3T3/virologia , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/genética , Proteína Ligante Fas , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/imunologia , Terapia Genética , Ligantes , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA