Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Ann Oncol ; 25(6): 1116-21, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24685829

RESUMO

BACKGROUND: Results from the phase III trial CLEOPATRA in human epidermal growth factor receptor 2-positive first-line metastatic breast cancer demonstrated significant improvements in progression-free and overall survival with pertuzumab, trastuzumab, and docetaxel over placebo, trastuzumab, and docetaxel. We carried out exploratory analyses of the incidence and time to development of central nervous system (CNS) metastases in patients from CLEOPATRA. PATIENTS AND METHODS: Patients received pertuzumab/placebo: 840 mg in cycle 1, then 420 mg; trastuzumab: 8 mg/kg in cycle 1, then 6 mg/kg; docetaxel: initiated at 75 mg/m(2). Study drugs were administered i.v. every 3 weeks. The log-rank test was used for between-arm comparisons of time to CNS metastases as first site of disease progression and overall survival in patients with CNS metastases as first site of disease progression. The Kaplan-Meier approach was used to estimate median time to CNS metastases as first site of disease progression and median overall survival. RESULTS: The incidence of CNS metastases as first site of disease progression was similar between arms; placebo arm: 51 of 406 (12.6%), pertuzumab arm: 55 of 402 (13.7%). Median time to development of CNS metastases as first site of disease progression was 11.9 months in the placebo arm and 15.0 months in the pertuzumab arm; hazard ratio (HR) = 0.58, 95% confidence interval (CI) 0.39-0.85, P = 0.0049. Overall survival in patients who developed CNS metastases as first site of disease progression showed a trend in favor of pertuzumab, trastuzumab, and docetaxel; HR = 0.66, 95% CI 0.39-1.11. Median overall survival was 26.3 versus 34.4 months in the placebo and pertuzumab arms, respectively. Treatment comparison of the survival curves was not statistically significant for the log-rank test (P = 0.1139), but significant for the Wilcoxon test (P = 0.0449). CONCLUSIONS: While the incidence of CNS metastases was similar between arms, our results suggest that pertuzumab, trastuzumab, and docetaxel delays the onset of CNS disease compared with placebo, trastuzumab, and docetaxel. CLINICALTRIALSGOV: NCT00567190.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/patologia , Neoplasias do Sistema Nervoso Central/epidemiologia , Neoplasias do Sistema Nervoso Central/secundário , Adulto , Idoso , Anticorpos Monoclonais Humanizados/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Docetaxel , Método Duplo-Cego , Feminino , Humanos , Incidência , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Receptor ErbB-2/genética , Taxoides/administração & dosagem , Trastuzumab
2.
Poult Sci ; 89(10): 2083-91, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20852098

RESUMO

Marek's disease (MD) is a T-cell lymphoma disease of domestic chickens induced by MD virus (MDV), a naturally oncogenic and highly contagious cell-associated α-herpesvirus. Earlier reports have shown that the MHC haplotype as well as non-MHC genes are responsible for genetic resistance to MD. The MHC was also shown to affect efficiency of vaccine response. Using specific-pathogen-free chickens from a series of 19 recombinant congenic strains and their 2 progenitor lines (lines 6(3) and 7(2)), vaccine challenge experiments were conducted to examine the effect of host genetic variation on vaccine efficacy. The 21 inbred lines of White Leghorns share the same B*2 MHC haplotype and the genome of each recombinant congenic strain differs by a random 1/8 sample of the susceptible donor line (7(2)) genome. Chickens from each of the lines were divided into 2 groups. One was vaccinated with turkey herpesvirus strain FC126 at the day of hatch and the other was treated as a nonvaccinated control. Chickens of both groups were inoculated with a very virulent plus strain of MDV on the fifth day posthatch. Analyses of the MD data showed that the genetic line significantly influenced MD incidence and days of survival post-MDV infection after vaccination of chickens (P<0.01). The protective indices against MD varied greatly among the lines with a range of 0 up to 84%. This is the first evidence that non-MHC host genetic variation significantly affects MD vaccine efficacy in chickens in a designed prospective study.


Assuntos
Galinhas/genética , Galinhas/imunologia , Variação Genética , Vacinas contra Doença de Marek/imunologia , Doença de Marek/prevenção & controle , Animais , Doença de Marek/imunologia , Organismos Livres de Patógenos Específicos
3.
Virology ; 379(2): 256-65, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18675437

RESUMO

The bursa of Fabricius serves as an important tissue in the process of Marek's disease virus (MDV) pathogenesis, since B cells of the bursa harbor the cytolytic phase of MDV replication cycle. In the present study, host responses associated with MDV infection in the bursa of Fabricius of chickens were investigated. The expression of MDV phosphoprotein (pp)38 antigen, MDV glycoprotein (gB) and MDV viral interleukin (vIL)-8 transcripts was at the highest at 4 days post-infection (d.p.i.) and then showed a declining trend. On the contrary, the expression of meq (MDV EcoRI Q) gene as well as the viral genome load increased gradually until day 14 post-infection. The changes in viral parameters were associated with significantly higher infiltration of macrophages and T cell subsets, particularly CD4+ T cells into the bursa of Fabricius. Of the genes examined, the expression of interferon (IFN)-alpha, IFN-gamma genes and inducible nitric oxide synthase (iNOS) was significantly up-regulated in response to MDV infection in the bursa of Fabricius. The results suggest a role for these cells and cytokines in MDV-induced responses in the bursa of Fabricius.


Assuntos
Bolsa de Fabricius/virologia , Mardivirus/patogenicidade , Animais , Animais Recém-Nascidos , Antígenos Virais/metabolismo , Sequência de Bases , Bolsa de Fabricius/imunologia , Bolsa de Fabricius/patologia , Galinhas , Citocinas/genética , Primers do DNA/genética , Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/patologia , Macrófagos/virologia , Mardivirus/genética , Mardivirus/imunologia , Mardivirus/fisiologia , Doença de Marek/genética , Doença de Marek/imunologia , Doença de Marek/patologia , Doença de Marek/virologia , Óxido Nítrico Sintase Tipo II/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia , Virulência , Replicação Viral
4.
Avian Pathol ; 37(1): 7-13, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202944

RESUMO

Immunohistochemistry and polymerase chain reaction (PCR) were used to test for the presence of avian leukosis virus (ALV) J viral antigen gp85 and proviral DNA, respectively, in various tissues (adrenal gland, bone marrow, gonad, heart, kidney, liver, lung, pancreas, proventriculus, sciatic nerve, spleen, and thymus). Tissues were collected from 32-week-old commercial meat-type and Avian Disease and Oncology Laboratory experimental White Leghorn Line 0 chickens with the following different infection profiles: tV + A-, included in ovo-tolerized viraemic chickens with no neutralizing antibodies (NAbs) on any sampling; ntV + A-, included chickens that were viraemic and NAb-negative at the time of termination at 32 weeks post hatch, but had NAbs on up to two occasions; V+ A+, included chickens that were viraemic and NAb-positive at the time of termination at 32 weeks post hatch, and had NAbs on more than two occasions; V - A+, included chickens that were negative for viraemia and NAb-positive at the time of termination at 32 weeks post hatch, and had antibody on more than two occasions; V - A-, included chickens that were never exposed to ALV J virus. There was a direct correlation between viraemia and tissue distribution of gp85, regardless of the NAb status and strain of chickens, as expression of ALV J gp85 was noted in only viraemic chickens (tV + A-, ntV + A-, V+ A+), but not in non-viraemic seroconverted chickens (V - A+). Of the four oligonucleotide primers pairs used in PCR to identify ALV J provirus, only one primer set termed H5/H7 was useful in demonstrating ALV J proviral DNA in the majority of the tissues tested from non-viraemic, antibody-positive chickens (V - A+). The results suggest that PCR using primer pair H5/H7 is more sensitive than immunohistochemistry in identifying ALV J in chickens that have been exposed to virus, but are not actively viraemic.


Assuntos
Antígenos Virais/isolamento & purificação , Vírus da Leucose Aviária/classificação , Leucose Aviária/virologia , Galinhas/genética , Provírus/isolamento & purificação , Animais , Antígenos Virais/metabolismo , Leucose Aviária/metabolismo , DNA Viral/isolamento & purificação , Carne , Estudos Retrospectivos
5.
J Vet Med A Physiol Pathol Clin Med ; 54(10): 553-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18045338

RESUMO

Avian leukosis virus (ALV) subgroup J (ALV-J) is an exogenous ALV and causes myeloid leukosis in meat-type chickens. We have previously reported the isolation and identification of ALV-J in commercial layer flocks from 12 farms in northern China. In this report, we further characterized this virus by in situ polymerase chain reaction (PCR) hybridization in various affected organs of chickens from six of the 12 farms. A routine method for hybridization of nucleic acid uses radioactive probe, such as a P32-labelled probe. We found that the non-radioactive digoxigenin (DIG) probe is sensitive enough to detect the nucleic acid of virus in chicken tissues. We used a pair of published primers (H5/H7) specific to the gp85 envelope gene and 3' region of pol gene of prototype ALV-J strain HPRS-103. The total RNA extracted from tumour, bone marrow, oviduct, liver and spleen of the diseased chickens from six commercial flocks, and cDNA was successfully amplified. Using the primers and cDNA, we obtained an ALV-J-specific cDNA probe of 545 bp in length by PCR. In situ PCR with H5/H7 primers was carried out in the paraffin sections from tissues of the diseased chickens, followed by in situ hybridization using the DIG-labelled cDNA probe. Positive hybridization signals were detected in the cytoplasm of paraffin sections of tumours and other organ tissues. The intensity of the signals was documented using an image analysis system measuring integral optical density (IOD). The IOD values for tissue sections treated by in situ PCR hybridization are significantly higher than that by in situ hybridization alone (P < 0.01). These data taken together suggest that in situ PCR hybridization is a more sensitive technique for detection of ALV-J in tissue sections.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/transmissão , Leucose Aviária/virologia , Doenças das Aves Domésticas/virologia , Animais , Galinhas , China , Feminino , Hibridização In Situ/veterinária , Rim/patologia , Rim/virologia , Fígado/patologia , Fígado/virologia , Oviductos/patologia , Oviductos/virologia , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/transmissão
6.
Avian Dis ; 51(3): 713-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17992931

RESUMO

Marek's disease virus (MDV) is an oncogenic cell-associated herpesvirus that causes T-cell lymphoma in chickens. Lymphoproliferative neoplasms in Marek's disease (MD) occur in various organs and tissues, including the viscera, peripheral nerves, skin, gonads, and musculatures. MDV is restrictively produced in the feather follicle epithelial (FFE) cells, and it gains access to the external environment via infected cells or as infectious enveloped cell-free virus particles. The goals of the present study were to 1) determine whether the MDV-induced skin lesions are neoplastic in nature or inflammatory reactions to viral infection, 2) determine whether physical presence of feather follicles (FF) is necessary for skin tumor development, and 3) study the role of skin epithelial cells not associated with feathers or FF in the replication and dissemination of infectious virus particles. Scaleless chickens that produce only a few scattered feathers and no sculate scales along the anterior metatarsi were used as a unique model to study the pathogenesis of dermal lesions. Histologic and immunohistochemical analysis revealed that the cutaneous lesions were tumorous as was manifested by massive accumulation of lymphoblasts and extensive activation of meq oncoprotein, the hallmark of MDV oncogenesis, within the skin lesions. Neoplastic cutaneous lesions in the scaleless chickens indicate that feather follicles are not necessary for skin tumor development. Finally, our preliminary data indicate that inoculation with supernatant fluid from homogenized and sonicated skin samples of MDV-infected scaleless chickens induces MD in susceptible birds, suggesting that skin epithelial cells not associated with FF also harbor infectious viral particles.


Assuntos
Galinhas/virologia , Plumas , Doença de Marek/patologia , Neoplasias Cutâneas/veterinária , Pele/patologia , Pele/virologia , Animais , Feminino , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/virologia
7.
J Virol ; 79(7): 4545-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15767457

RESUMO

Marek's disease virus has a unique phosphoprotein, pp38, which is suspected to play an important role in Marek's disease pathogenesis. The objective of the present study was to utilize a mutant virus lacking the pp38 gene (rMd5Deltapp38) to better characterize the biological function of pp38. This work shows that the pp38 gene is necessary to establish cytolytic infection in B cells but not in feather follicle epithelium, to produce an adequate level of latently infected T cells, and to maintain the transformed status in vivo.


Assuntos
Antígenos Virais/fisiologia , Linfócitos B/virologia , Transformação Celular Viral/fisiologia , Células Epiteliais/virologia , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/fisiologia , Fosfoproteínas/fisiologia , Animais , Antígenos Virais/genética , Galinhas , Deleção de Genes , Genes Virais , Fosfoproteínas/genética
8.
Proc Natl Acad Sci U S A ; 101(38): 13879-84, 2004 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-15356338

RESUMO

Animal models are essential for elucidating the molecular mechanisms of carcinogenesis. Hodgkin's and many diverse non-Hodgkin's lymphomas overexpress the Hodgkin's disease antigen CD30 (CD30(hi)), a tumor necrosis factor receptor II family member. Here we show that chicken Marek's disease (MD) lymphoma cells are also CD30(hi) and are a unique natural model for CD30(hi) lymphoma. Chicken CD30 resembles an ancestral form, and we identify a previously undescribed potential cytoplasmic signaling domain conserved in chicken, human, and mouse CD30. Our phylogeneic analysis defines a relationship between the structures of human and mouse CD30 and confirms that mouse CD30 represents the ancestral mammalian gene structure. CD30 expression by MD virus (MDV)-transformed lymphocytes correlates with expression of the MDV Meq putative oncogene (a c-Jun homologue) in vivo. The chicken CD30 promoter has 15 predicted high-stringency Meq-binding transcription factor recognition motifs, and Meq enhances transcription from the CD30 promoter in vitro. Plasma proteomics identified a soluble form of CD30. CD30 overexpression is evolutionarily conserved and defines one class of neoplastic transformation events, regardless of etiology. We propose that CD30 is a component of a critical intracellular signaling pathway perturbed in neoplastic transformation. Specific anti-CD30 Igs occurred after infection of genetically MD-resistant chickens with oncogenic MDV, suggesting immunity to CD30 could play a role in MD lymphoma regression.


Assuntos
Doença de Hodgkin/genética , Antígeno Ki-1/genética , Mardivirus/imunologia , Doença de Marek/genética , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Transformação Celular Neoplásica/imunologia , Galinhas , Sequência Conservada , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/imunologia , Doença de Hodgkin/imunologia , Humanos , Ativação Linfocitária/imunologia , Doença de Marek/imunologia , Camundongos , Dados de Sequência Molecular , Peso Molecular , Filogenia , Alinhamento de Sequência , Transdução de Sinais/imunologia
9.
Avian Pathol ; 33(1): 13-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14681063

RESUMO

Mortality from myeloid leukosis was observed in commercial layers from 12 farms in northern China. Affected chickens were extremely thin and dehydrated, bleeding occurred in feather follicles and claws, combs were pale and anaemic, phalanges were swollen, and many yellowish-white tumours were seen on the visceral surface of the sternum. Focal tumour cells, with spherical eosinophilic granules in the cytoplasm, were found in the liver, spleen, kidney, ovary, oviduct, lung, bone marrow, proventriculus and gut by histopathological examination. Immunohistochemical studies with a monoclonal antibody to gp85 of avian leukosis virus subgroup J (ALV-J) revealed antigen in all organs examined. Polymerase chain reaction tests using a pair of ALV-J-specific primers H5/H7 (Smith et al., 1998) produced a 545 basepair fragment. The sequence of the Polymerase chain reaction product was compared with that of the ALV-J HPRS-103 prototype strain. The identity of nucleotides and predicted amino acids was 97.4% and 96.1%, respectively. On this basis the disease in the egg-type chickens was diagnosed as an ALV-J infection. This is the first report of field cases of myeloid leukosis caused by ALV-J in commercial egg-type chickens.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/epidemiologia , Galinhas , Doenças das Aves Domésticas/epidemiologia , Animais , Antígenos Virais/análise , Leucose Aviária/patologia , Leucose Aviária/virologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/imunologia , Sequência de Bases , China/epidemiologia , Feminino , Imuno-Histoquímica/veterinária , Dados de Sequência Molecular , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Homologia de Sequência do Ácido Nucleico
10.
Mol Cell Biol ; 21(24): 8385-97, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11713275

RESUMO

The bombesin/gastrin-releasing peptide (GRP) family of neuropeptides has been implicated in various in vitro and in vivo models of human malignancies including prostate cancers. It was previously shown that bombesin and/or neurotensin (NT) acts as a survival and migratory factor(s) for androgen-independent prostate cancers. However, a role in the transition from an androgen-dependent to -refractory state has not been addressed. In this study, we investigate the biological effects and signal pathways of bombesin and NT on LNCaP, a prostate cancer cell line which requires androgen for growth. We show that both neurotrophic factors can induce LNCaP growth in the absence of androgen. Concurrent transactivation of reporter genes driven by the prostate-specific antigen promoter or a promoter carrying an androgen-responsive element (ARE) indicate that growth stimulation is accompanied by androgen receptor (AR) activation. Furthermore, neurotrophic factor-induced gene activation was also present in PC3 cells transfected with the AR but not in the parental line which lacks the AR. Given that bombesin does not directly bind to the AR and is known to engage a G-protein-coupled receptor, we investigated downstream signaling events that could possibly interact with the AR pathway. We found that three nonreceptor tyrosine kinases, focal adhesion kinase (FAK), Src, and Etk/BMX play important parts in this process. Etk/Bmx activation requires FAK and Src and is critical for neurotrophic factor-induced growth, as LNCaP cells transfected with a dominant-negative Etk/BMX fail to respond to bombesin. Etk's activation requires FAK, Src, but not phosphatidylinositol 3-kinase. Likewise, bombesin-induced AR activation is inhibited by the dominant-negative mutant of either Src or FAK. Thus, in addition to defining a new G-protein pathway, this report makes the following points regarding prostate cancer. (i) Neurotrophic factors can activate the AR, thus circumventing the normal growth inhibition caused by androgen ablation. (ii) Tyrosine kinases are involved in neurotrophic factor-mediated AR activation and, as such, may serve as targets of future therapeutics, to be used in conjunction with current antihormone and antineuropeptide therapies.


Assuntos
Androgênios/metabolismo , Neuropeptídeos/metabolismo , Proteínas Tirosina Quinases/fisiologia , Quinases da Família src/fisiologia , Bombesina/metabolismo , Bombesina/farmacologia , Bromodesoxiuridina/metabolismo , Divisão Celular , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Genes Dominantes , Genes Reporter , Humanos , Luciferases/metabolismo , Masculino , Modelos Biológicos , Plasmídeos/metabolismo , Testes de Precipitina , Neoplasias da Próstata/metabolismo , Ligação Proteica , Transdução de Sinais , Fatores de Tempo , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
11.
Arch Virol ; 146(5): 983-92, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11448035

RESUMO

Glycoproteins H and L form a hetero-oligomeric complex (gH-L) which plays an important role in virus entry to host cells and cell-to-cell infection in herpesviruses. Interaction of gH and gL is considered to be critical for the biological function of these two glycoproteins. To investigate the interaction of MDV gH and gL, both gH and gL were expressed in in vitro cell culture systems using indirect immunofluorescence assay with gH and gL antibodies. The results suggested that co-expression of gH and gL in the same cells are required and necessary for both gH and gL subcellular translocation and cell surface expression. gL expressed in recombinant fowlpox virus (rFPV) infected chicken embryo fibroblasts (CEF) was consistently secreted into the culture medium. The primary peptide of gL binds with that of gH in the cytosol or ER lumen. By binding with gH, gL could anchor itself on the cell surface allowing for surface expression and viral spread to uninfected cells. The binding domain of gH was mapped to the amino acids 451-659 (SacI-HindIII) fragment and was essential for gH-L complex formation.


Assuntos
Herpesvirus Galináceo 2/fisiologia , Proteínas do Envelope Viral/fisiologia , Animais , Anticorpos Antivirais/imunologia , Sítios de Ligação , Biopolímeros , Células Cultivadas/virologia , Embrião de Galinha , Meios de Cultivo Condicionados , Fibroblastos/virologia , Técnica Indireta de Fluorescência para Anticorpo , Vetores Genéticos/genética , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/imunologia , Substâncias Macromoleculares , Nucleopoliedrovírus/genética , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/fisiologia , Spodoptera , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Cultura de Vírus
12.
J Virol ; 75(11): 5159-73, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333897

RESUMO

Chemokines induce chemotaxis, cell migration, and inflammatory responses. We report the identification of an interleukin-8 (IL-8) homolog, termed vIL-8, encoded within the genome of Marek's disease virus (MDV). The 134-amino-acid vIL-8 shares closest homology to mammalian and avian IL-8, molecules representing the prototype CXC chemokine. The gene for vIL-8 consists of three exons which map to the BamHI-L fragment within the repeats flanking the unique long region of the MDV genome. A 0.7-kb transcript encoding vIL-8 was detected in an n-butyrate-treated, MDV-transformed T-lymphoblastoid cell line, MSB-1. This induction is essentially abolished by cycloheximide and herpesvirus DNA polymerase inhibitor phosphonoacetate, indicating that vIL-8 is expressed with true late (gamma2) kinetics. Baculovirus-expressed vIL-8 was found to be secreted into the medium and shown to be functional as a chemoattractant for chicken peripheral blood mononuclear cells but not for heterophils. To characterize the function of vIL-8 with respect to MDV infection in vivo, a recombinant MDV was constructed with a deletion of all three exons and a soluble-modified green fluorescent protein (smGFP) expression cassette inserted at the site of deletion. In two in vivo experiments, the vIL-8 deletion mutant (RB1BvIL-8DeltasmGFP) showed a decreased level of lytic infection in comparison to its parent virus, an equal-passage-level parent virus, and to another recombinant MDV containing the insertion of a GFP expression cassette at the nonessential US2 gene. RB1BvIL-8DeltasmGFP retained oncogenicity, albeit at a greatly reduced level. Nonetheless, we have been able to establish a lymphoblastoid cell line from an RB1BvIL-8DeltasmGFP-induced ovarian lymphoma (MDCC-UA20) and verify the presence of a latent MDV genome lacking vIL-8. Taken together, these data describe the identification and characterization of a chemokine homolog encoded within the MDV genome that is dispensable for transformation but may affect the level of MDV in vivo lytic infection.


Assuntos
Fatores Quimiotáticos/genética , Herpesvirus Galináceo 2/imunologia , Interleucina-8/biossíntese , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Baculoviridae/genética , Sequência de Bases , Linhagem Celular , Linhagem Celular Transformada , Galinhas , Clonagem Molecular , Cicloeximida , Deleção de Genes , Proteínas de Fluorescência Verde , Herpesvirus Galináceo 2/genética , Interleucina-8/genética , Leucócitos Mononucleares/metabolismo , Proteínas Luminescentes/genética , Dados de Sequência Molecular , Mutagênese Insercional , Ácido Fosfonoacéticos , RNA Mensageiro/análise , Proteínas Recombinantes/biossíntese , Alinhamento de Sequência , Proteínas Virais/biossíntese , Proteínas Virais/genética , Replicação Viral
13.
Virology ; 282(1): 198-205, 2001 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-11259202

RESUMO

Infection of chicken cells with three Marek's disease virus (MDV) serotypes interferes with expression of the major histocompatibility complex (MHC or B complex) class I (BF) glycoproteins. BF surface expression is blocked after infection of OU2 cells with MDV serotypes 1, 2, and 3. MDV-induced T-cell tumors suffer a nearly complete loss of cell surface BF upon virus reactivation with 5-bromo-2'-deoxyuridine (BUdR). The recombinant virus (RB1BUS2gfpDelta) transforming the MDCC-UA04 cell line expresses green fluorescent protein (GFP) during the immediate early phase of viral gene expression. Of the UA04 cells induced to express the immediate early GFP, approximately 60% have reduced levels of BF expression. All of the reactivated UA04 and MSB1 tumor cells expressing the major early viral protein pp38 display reduced levels of BF. Thus, BF down-regulation begins in the immediate early phase and is complete by the early phase of viral gene expression. The intracellular pool of BF is not appreciably affected, indicating that the likely mechanism is a block in BF transport and not the result of transcriptional or translational regulation.


Assuntos
Membrana Celular/metabolismo , Herpesvirus Galináceo 2/patogenicidade , Antígenos de Histocompatibilidade Classe I/metabolismo , Animais , Western Blotting , Linhagem Celular , Galinhas , Regulação para Baixo , Citometria de Fluxo , Herpesvirus Galináceo 2/genética
16.
Avian Dis ; 45(4): 938-45, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11785897

RESUMO

In an attempt to develop a specific diagnostic test for avian leukosis virus (ALV) subgroup J (ALV-J) strain Hc1, four monoclonal antibodies (MAbs), JE9, G2, 145, and J47, were generated that are specific for ALV-J envelope glycoprotein, gp85. Polymerase chain reaction (PCR) was used to amplify genomic pro-viral DNA of Avian Disease and Oncology Laboratory (ADOL)-Hc1 and ADOL-4817 envelope genes. Both open reading frames encoding glycoproteins gp85 and gp37 were cloned into baculoviruses. Abundant expression of gp85 and gp37 was detected in the recombinant viruses with specific antibody to Hc1 strain of the ALV-J. The expressed proteins were used for immunization of mice to produce hybridoma cell lines secreting MAbs specific to ALV-J envelope protein. A panel of MAbs was generated by fusing NS1 myeloma cells and spleen cells from mice immunized with the recombinant baculoviruses. With the use of an immunofluorescence assay, three MAbs (JE9, G2, 145) reacted with ALV-J but not with subgroups A, B, C, D, or E of ALV. MAb J47 reacted with all exogenous subgroups of ALV including A, B, C, D, and J but not with endogenous subgroup E viruses. Western blot analysis was performed with all four MAbs against recombinant baculovirus and Hc1-infected chicken embryo fibroblast (CEF) lysates. A major band with a molecular weight about 90 kD corresponding to the size of ALV-J envelope was consistently obtained. With these MAbs, we detected the Hc1 antigen in CEFs infected with several ALV-J viruses isolated in the United States and also in tissue sections from chickens infected with Hc1 strain of ALV-J. These MAbs will be useful reagents for the diagnosis of ALV-J infection because they recognize a common antigenic epitope in six isolates tested thus far.


Assuntos
Anticorpos Monoclonais/imunologia , Vírus da Leucose Aviária/imunologia , Leucose Aviária/diagnóstico , Galinhas , Animais , Anticorpos Monoclonais/análise , Especificidade de Anticorpos , Leucose Aviária/imunologia , Vírus da Leucose Aviária/classificação , Western Blotting/veterinária , Células Cultivadas , DNA Viral , Técnica Indireta de Fluorescência para Anticorpo/veterinária , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Peso Molecular , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase/veterinária , Sensibilidade e Especificidade , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
17.
Cancer Immunol Immunother ; 49(2): 78-84, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10823417

RESUMO

Lung cancer is a leading cause of cancer-related death in the United States. For this reason we chose to study the specific cellular effects that one chemotherapeutic agent, paclitaxel, has on lung carcinoma. In addition to its known mechanism of action, which is to stabilize microtubules, paclitaxel has been shown to have other interesting and relevant cellular effects. In this report, we demonstrate that a subset of human lung carcinoma cell lines respond to paclitaxel treatment with an up to a fivefold increase in the production of interleukin-8 (IL-8). We demonstrate that this increased production is specific to IL-8 but not to other chemokines, and is both dose- and time-dependent. Increased IL-8 mRNA is seen as early as 45 min with a peak at 4 h after paclitaxel treatment. This increase in mRNA is due to transcriptional activation because actinomycin D treatment blocked the increase. Paclitaxel also activates the mitogen-activated protein kinase family member, JNK1, in dose-dependent fashion. IL-8 enhancement is completely abolished with the use of an inhibitor of NF-kappaB, the super-repressor IkappaB. Similar results were obtained upon the inhibition of AP-1 activation with the MEK1/2 inhibitor, U0126. By gaining a better understanding of the differences in cellular response to paclitaxel chemotherapy, these findings might lead to either improved patient selection or to the development of adjuvant therapy targeted at specific-cell signaling proteins.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Interleucina-8/biossíntese , Neoplasias Pulmonares/tratamento farmacológico , NF-kappa B/fisiologia , Paclitaxel/farmacologia , Fator de Transcrição AP-1/fisiologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Humanos , Neoplasias Pulmonares/imunologia , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Tumorais Cultivadas , Regulação para Cima
18.
Proc Natl Acad Sci U S A ; 97(11): 6091-6, 2000 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-10823954

RESUMO

We have determined the DNA sequence of the unique long (UL) region and the repeat long (RL) region in the genome of serotype 1 GA strain of Marek's disease virus (MDV), a member of the alpha-herpesvirus family. With this information, the complete nucleotide sequence of GA-MDV is now known. The entire GA-MDV genome is predicted to be about 174 kbp in size, with an organization of TRL-UL-IRL-IRS-US-TRS, typical of a alpha-herpesvirus. The UL sequence contains 113,508 bp and has a base composition of 41.7% G + C. A total of 67 ORFs were identified completely within the UL region, among which 55 are homologous to genes encoded by herpes simplex virus-1. Twelve of them are unique with presently unknown functions. The sequence of RL reported here together with those published earlier reveal the major structural features of the RL. Virtually all of the ORFs encoded by RL are specific to serotype I of MDV. These ORFs are likely to contribute to some of the unique biological properties of MDV. Among the proteins encoded by MDV-specific ORFs are Meq, a jun/fos family of transcriptional factor implicated in transformation and latency, virus-encoded interleukin-8, a CXC chemokine, and pp38 and pp24, two phosphoproteins with undefined functions. There is also a putative lipase gene (LORF2) that has homologies in HPRS-24 (serotype II) strain of MDV and in various avian adenoviruses. An additional unique feature of MDV is the presence of long terminal repeat remnant sequences of avian retrovirus reticuloendotheliosis virus. These remnant sequences are derived from the U3-enhancer region through ancestral insertions by reticuloendotheliosis virus proviruses.


Assuntos
DNA Viral/genética , Genoma Viral , Herpesvirus Galináceo 2/genética , Replicação do DNA/genética , Glicoproteínas/genética , Herpesvirus Galináceo 2/classificação , Dados de Sequência Molecular , Fases de Leitura Aberta , Análise de Sequência de DNA , Sorotipagem , Especificidade da Espécie , Proteínas Virais/genética , Vírion/genética , Replicação Viral/genética
19.
J Immunol ; 164(5): 2769-75, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10679119

RESUMO

Paclitaxel is a frontline therapy for ovarian cancer. Our laboratory has shown that paclitaxel induces IL-8, a member of the C-X-C family of chemokines, in subsets of human ovarian cancer cells. However, the critical issue concerns the biological significance of this chemokine in human ovarian cancer. To study the influence of IL-8 on tumor growth, human ovarian cancer cell lines were transfected with an expression vector for human IL-8 and tested for their ability to form tumors in nude mice. IL-8 expression by the transfected cells did not alter their growth properties in vitro. In contrast, tumor growth in vivo was significantly attenuated in animals receiving IL-8-expressing cells when compared with mice injected with control cells. As additional evidence that IL-8 is a crucial factor in tumor growth, it was noted that ovarian cell lines in which constitutive IL-8 expression is elevated did not form tumors. Injection of neutralizing Ab to IL-8 reverted the phenotype and caused tumor growth in vivo. Examination of tissue from the inoculation site revealed a dramatically elevated cellularity, containing neutrophils and macrophages, in mice receiving IL-8-expressing tumor cells. These results suggest that IL-8 production by human ovarian tumor cells can play a role in reducing the rate of tumor growth; this effect may be mediated by the increased targeting of neutrophil and other mononuclear cells to the tumor injection site. These studies indicate a role for IL-8 in ovarian cancer control and suggest that chemotherapy-induced IL-8 may have a positive role in controlling tumor growth.


Assuntos
Movimento Celular/imunologia , Interleucina-8/fisiologia , Neutrófilos/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/prevenção & controle , Animais , Divisão Celular/imunologia , Modelos Animais de Doenças , Feminino , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/fisiologia , Humanos , Soros Imunes/administração & dosagem , Imuno-Histoquímica , Interleucina-8/biossíntese , Interleucina-8/imunologia , Leucócitos Mononucleares/química , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Monócitos/química , Monócitos/imunologia , Monócitos/patologia , Transplante de Neoplasias , Neutrófilos/química , Neutrófilos/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Coloração e Rotulagem , Células Tumorais Cultivadas
20.
Virology ; 264(1): 205-10, 1999 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-10544146

RESUMO

A cell line (DF-1¿J) expressing the envelope protein isolated from the ADOL-Hc1 strain of the avian leukosis virus subgroup J (ALV-J) was used to analyze receptor interference to six different isolates of ALV-J as well as ALV subgroups A-D. The traditional gag-specific enzyme-linked immunosorbent assay (ELISA) as well as flow cytometry was used to evaluate viral infection. The parental cell line (DF-1) was susceptible to all ALV subgroups tested while the DF-1¿J cell line was selectively resistant to the subgroup J isolates. The DF-1¿J cell line was resistant to infection by all six ALV-J isolates as determined using the gag-specific ELISA. There was no interference with the other ALV subgroups (A-D) induced by the expression of the ADOL-Hcl envelope. The ALV-J isolates used in this analysis are serologically distinct when analyzed by flow cytometry. Convalescent sera to ADOL-Hcl cross-reacts with all of the ALV-J isolates tested; however, sera to HPRS-103 did not bind to four of the six isolates. Based on the intensity and differential binding of these antisera using flow cytometry, the six ALV-J isolates used can be grouped into four categories. Thus the DF-1¿J cell line is resistant to infection by a serologically and genetically diverse group of ALV-J isolates and should be useful as a diagnostic tool.


Assuntos
Vírus da Leucose Aviária/fisiologia , Vírus da Leucose Aviária/patogenicidade , Engenharia Genética , Receptores Virais/fisiologia , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Embrião de Galinha , Reações Cruzadas , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Fibroblastos , Citometria de Fluxo , Genes gag , Soros Imunes , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA