Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 185(5): 2895-902, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20675595

RESUMO

T-bet (Tbx21), a T-box transcription factor, has been previously identified as a master regulator of type 1 T cell polarization. We have also recently shown that the genetic engineering of human dendritic cells (DCs) to express human T-bet cDNA yields type 1-polarizing APCs in vitro (1). In the present study, murine CD11c(+) DCs were transduced with a recombinant adenovirus encoding full-length murine T-bets (DC.mTbets) and analyzed for their immunomodulatory functions in vitro and in vivo. Within the range of markers analyzed, DC.mTbets exhibited a control DC phenotype and were indistinguishable from control DCs in their ability to promote allogenic T cell proliferation in MLR in vitro. However, DC.mTbets were superior to control DCs in promoting Th1 and Tc1 responses in vitro via a mechanism requiring DC-T cell interaction or the close proximity of these two cell types and that can only partially be explained by the action of DC-elaborated IL-12p70. When injected into day 7 s.c. CMS4 sarcoma lesions growing in syngenic BALB/c mice, DC.mTbets dramatically slowed tumor progression (versus control DCs) and extended overall survival via a mechanism dependent on both CD4(+) and CD8(+) T cells and, to a lesser extent, asialoGM1(+) NK cells. DC.mTbet-based therapy also promoted superior tumor-specific Tc1 responses in the spleens and tumor-draining lymph nodes of treated animals, and within the tumor microenvironment it inhibited the accumulation of CD11b(+)Gr1(+) myeloid-derived suppressor cells and normalized CD31(+) vascular structures. These findings support the potential translational utility of DC.Tbets as a therapeutic modality in the cancer setting.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Engenharia Genética/métodos , Imunidade Inata/genética , Injeções Intralesionais/métodos , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Proteínas com Domínio T/administração & dosagem , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica/imunologia , Antígenos H-2/administração & dosagem , Antígenos H-2/genética , Humanos , Imunofenotipagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sarcoma Experimental/mortalidade , Sarcoma Experimental/virologia , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Transdução Genética
2.
J Gen Virol ; 91(Pt 9): 2176-85, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20573854

RESUMO

Cancers with viral aetiologies can potentially be prevented by antiviral vaccines. Therefore, it is important to understand how viral infections and cancers might be linked. Some cancers frequently carry gammaherpesvirus genomes. However, they generally express the same viral genes as non-transformed cells, and differ mainly in also carrying oncogenic host mutations. Infection, therefore, seems to play a triggering or accessory role in disease. The hit-and-run hypothesis proposes that cumulative host mutations can allow viral genomes to be lost entirely, such that cancers remaining virus-positive represent only a fraction of those to which infection contributes. This would have considerable implications for disease control. However, the hit-and-run hypothesis has so far lacked experimental support. Here, we tested it by using Cre-lox recombination to trigger transforming mutations in virus-infected cells. Thus, 'floxed' oncogene mice were infected with Cre recombinase-positive murid herpesvirus-4 (MuHV-4). The emerging cancers showed the expected genetic changes but, by the time of presentation, almost all lacked viral genomes. Vaccination with a non-persistent MuHV-4 mutant nonetheless conferred complete protection. Equivalent human gammaherpesvirus vaccines could therefore potentially prevent not only viral genome-positive cancers, but possibly also some cancers less suspected of a viral origin because of viral genome loss.


Assuntos
Vacinas Anticâncer/farmacologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Rhadinovirus/imunologia , Rhadinovirus/patogenicidade , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Vacinas Virais/farmacologia , Animais , Sequência de Bases , Primers do DNA/genética , Genes p53 , Genes ras , Genoma Viral , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Modelos Biológicos , Mutagênese Insercional , Mutação , Rhadinovirus/genética , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Sarcoma Experimental/prevenção & controle , Sarcoma Experimental/virologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/virologia , Vacinação/métodos
3.
Folia Biol (Krakow) ; 58(1-2): 9-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20420189

RESUMO

Blood cell counts, differential blood cell counts and weights of the spleen and peripheral lymph nodes draining the area of lesions induced by Moloney sarcoma virus inoculation into the quadriceps shank muscles of inbred BALB/c mice were examined at various stages of tumor development and regression. The blood cell count remained constant through the observation period up to 27 days post tumor development and regression. Differential counts revealed some changes in the cellular composition of the peripheral blood. The most pronounced was an increase of neutrophiles at the stage of tumor development, and their decline with tumour regression. The enlargement of the spleen and of the popliteal lymph nodes draining the tumour site, at the peak of tumor development on day 13 post MSV inoculation, involved at least a doubling of splenic weight, and a much greater weight increase for lymph nodes. This was a long-lasting, although declining event, extending beyond tumor regression.


Assuntos
Tecido Linfoide/patologia , Vírus do Sarcoma Murino de Moloney , Infecções por Retroviridae/sangue , Sarcoma Experimental/sangue , Infecções Tumorais por Vírus/sangue , Animais , Contagem de Células Sanguíneas , Feminino , Tecido Linfoide/virologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/patologia , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Sarcoma Experimental/patologia , Sarcoma Experimental/virologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
4.
Leukemia ; 9 Suppl 1: S89-92, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7475323

RESUMO

In sarcomagenesis in rats infected neonatally with feline sarcoma virus (ST-FeSV), v-fes product (P85) was previously shown by us to be a predictive and preventive determinant. In order to explore the part played by P85 in tumor suppression, DNA was extracted from precancerous granulomas and from slow or rapid growing sarcomas induced by neonatal injection of the virus. The v-fes signal from extracted DNA was analyzed by PCR-SSCP. The prototype v-fes gene signal was detected in most lesions and found to be generally amplified in rapid growing sarcomas and in some granulomas. Several v-fes homologs showing varying mobilities in gel were seen in most sarcomas and some granulomas with or without the prototype v-fes signal. In slow growing sarcomas and granulomas induced in hosts that were immunized with ST-FeSV induced syngeneic sarcoma and proved to carry IgG antibody to P85, the prototype v-fes gene was found to be down-regulated and v-fes homologs were found to be reduced in number or eliminated. These results suggest that the development of v-fes mutations is associated with the growth potential of cells carrying the v-fes gene, and that host immunity to v-onc product influences the development of virogene rearrangements and results in slow and suppressed growth of tumors caused by neonatal infection with retrovirus.


Assuntos
Proteínas de Fusão gag-onc/biossíntese , Oncogenes , Vírus do Sarcoma Felino/patogenicidade , Sarcoma Experimental/genética , Sarcoma Experimental/patologia , Vacinas Virais , Animais , Animais Recém-Nascidos , Sequência de Bases , Gatos , Linhagem Celular , Primers do DNA , DNA Viral/análise , Feminino , Proteínas de Fusão gag-onc/genética , Granuloma/patologia , Granuloma/virologia , Hibridização In Situ , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Sinais Direcionadores de Proteínas/biossíntese , Ratos , Ratos Wistar , Vírus do Sarcoma Felino/genética , Vírus do Sarcoma Felino/isolamento & purificação , Sarcoma Experimental/prevenção & controle , Sarcoma Experimental/virologia
5.
Cancer Gene Ther ; 7(3): 422-9, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10766348

RESUMO

Mutations of the p53 tumor-suppressor gene are the most frequent genetic abnormality in soft tissue sarcomas. Because these rare tumors also respond poorly to standard chemotherapy and bear a 50% 5-year mortality rate, we investigated the possible therapeutic benefits of p53 gene restoration in sarcomas. We constructed Ad5p53, which is an E1A-deleted, replication-deficient adenovirus expressing a cytomegalovirus promoter-driven wild-type p53 cDNA with a Flag sequence tag. SKLMS-1 human leiomyosarcoma cells containing a mis-sense p53 point mutation were effectively transduced with Ad5p53. Increasing levels of Flag-p53 protein, as well as dose-dependent p21Cip1 induction, were observed through a dose range of 10-500 plaque-forming units (PFU)/cell. In vitro administration of Ad5p53 as a single 100 PFU/cell dose caused 40-60% growth inhibition of SKLMS-1 cells at posttreatment days 4, 6, and 8 compared with untreated or viral control treated-cells (P < .05, Student's t test). Relative to these same controls, in vivo treatment of SKLMS-1-bearing severe combined immunodeficient mice with 6 x 10(9) PFU of Ad5p53 by intratumoral injection resulted in a 35-day tumor growth delay and complete tumor regression in 40% of mice (P < .05, Student's t test). The expression of virally derived p53 mRNA in Ad5p53-treated tumor tissues was detected in treated tumor specimens by reverse transcriptase polymerase chain reaction. Reduced intratumoral cellularity and the presence of p53 staining in adjacent normal tissue, consistent with delivery of exogenous p53 to the tumor target, were evident only in Ad5p53-treated tumors after immunohistochemical staining for p53. These results indicate that wild-type p53 gene restoration in sarcomas retards tumor growth and may come to be usefully applied to the clinical treatment of this disease as a single regimen or in combination with conventional therapies.


Assuntos
Adenovírus Humanos/genética , Genes p53 , Terapia Genética/métodos , Sarcoma Experimental/genética , Sarcoma Experimental/terapia , Animais , Antineoplásicos/administração & dosagem , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/síntese química , Inibidores do Crescimento/administração & dosagem , Humanos , Injeções Intralesionais , Leiomiossarcoma/genética , Leiomiossarcoma/patologia , Leiomiossarcoma/terapia , Leiomiossarcoma/virologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Sarcoma Experimental/patologia , Sarcoma Experimental/virologia , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/farmacologia
6.
Transplantation ; 61(2): 292-9, 1996 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-8600639

RESUMO

Lymphocytotropic mouse parvoviruses can perturb immune responses. For example the recently identified mouse parvovirus designated MPV-1 persistently infects lymphoid tissues and interferes with the ability of cloned T cells to proliferate. As a consequence of these findings the present studies were undertaken to characterize further the immunomodulatory effects of MPV-1 on T cell-mediated immune responses in vivo and in vitro. To evaluate the effect of MPV-1 on CD8+ T cell-mediated responses sarcoma I (SaI) cells, devoid of class II major histocompatibility (MHC) antigens, were administered to MPV-1-infected adult BALB/c mice. MPV-1 infection accelerated tumor allograft rejection. Immunofluorescence staining and in situ hybridization studies of tumors suggested that direct infection of the tumor cells was not responsible for accelerated rejection. Furthermore, compared with uninfected mice, T cells from infected mice that had rejected SaI tumors had a diminished cytolytic capacity. Taken together these results suggest that MPV-1 may induce "bystander help." To examine the in vivo effect of MPV-1 on CD4+ T cell mediated responses adult mice were primed with ovalbumin (OVA) and infected with MPV-1. Spleen and popliteal lymph node cells from OVA-primed mice 3 or 7 days after MPV-1 inoculation had reduced proliferation responses, whereas the proliferative capacity of mesenteric lymph node cells from these mice was increased. Similarly, MPV-1 reduced cytokine-induced proliferation of allospecific CD8+ cloned L3 T cells and OVA-reactive CD4+ T cells without effecting cell viability. Since parvoviruses are widespread among laboratory rodents, these findings emphasize the importance of identifying and excluding parvovirus infections in mice used for transplantation studies and in cultures of mouse T lymphocytes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Rejeição de Enxerto/imunologia , Transplante de Neoplasias/imunologia , Infecções por Parvoviridae/imunologia , Parvovirus/imunologia , Animais , Rejeição de Enxerto/virologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Transplante de Neoplasias/patologia , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Sarcoma Experimental/virologia , Transplante Homólogo
7.
Anticancer Res ; 16(4A): 1835-42, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8712710

RESUMO

BK virus was isolated by Gardner et al in 1971 from the urine of an immunosuppressed patient with a kidney allograft. Antibodies to this virus are ubiquitous in the general populations worldwide, but the oncogenic capacity of BKV in humans had not been reported. The virus transformed in vitro permissive human and non-permissive animal cells, and the transformed cells had the T antigen. Intracerebral and intravenous inoculation of BKV in newborn hamsters induced malignant tumours (mainly ependymomas, malignant insulinomas, and osteosarcomas). Subcutaneous and intraperitoneal routes were also effective. The virus was only rescued from a few tumours by fusion with human embryonic cells or Vero cells. Brain tumours appeared earlier and osteosarcomas developed in animals which survived for more than 6 months. Many of the osteosarcomas were bony and grew slowly with frequent lung metastases, and a few osteosarcomas were soft and grew rapidly without lung metastases. Experimental targeting chemotherapy with doxorubicin (DX)-containing immunoliposomes was performed against Os515 osteosarcoma. In in vitro experiments, DX-Lip-MoAb29 showed a more significant inhibitory effect on cultured Os515 cells than free Dx and DX-Lip. DX-Lip DNP had less effect. In in vivo experiments, DX-Lip-MoAb29 suppressed the growth of Os515 tumour isografts in hamsters and prolonged the survival of recipients more significantly than free DX.


Assuntos
Vírus BK , Neoplasias Encefálicas/patologia , Osteossarcoma/patologia , Sarcoma Experimental/patologia , Animais , Animais Recém-Nascidos , Neoplasias Encefálicas/virologia , Chlorocebus aethiops , Cricetinae , Doxorrubicina/uso terapêutico , Embrião de Mamíferos , Ependimoma/patologia , Ependimoma/virologia , Humanos , Insulinoma/patologia , Insulinoma/virologia , Fusão de Membrana , Osteossarcoma/tratamento farmacológico , Osteossarcoma/virologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/virologia , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/virologia , Células Vero
8.
Hokkaido Igaku Zasshi ; 62(4): 581-7, 1987 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-2824311

RESUMO

Analysis based on the complement-dependent cytotoxicity (CDC) assays using syngeneic antiserum against a Rous sarcoma virus(RSV)-induced mouse tumor(CSA1M) showed that a cross-reactive antigen with a common tumor-associated cell surface antigen(TASA) of RSV-induced mouse tumors was shared with two human tumors A431 and MDA-468 overexpressing epidermal growth factor receptor(EGFR). The TASA, however, was not expressed on four human choriocarcinomas, a human lung cancer A2182, and human embryo fibroblasts HFF. Immunofluorescent studies demonstrated that A431 does not express a src gene product detected by anti-pp60src monoclonal antibody(MoAb). Two variant clones derived from A431 reducing number of ERFR (cl-15 and cl-16) have almost same growth rate and expression of transferrin receptor(Tf-R) in comparison with parental A431 cells. These clones, however, decreased the expression of TASA. Furthermore, CDC assays and enzyme-linked immunosorbent assay(ELISA) revealed that A431 reduced the expression of the TASA by pretreatment of EGF, but not with insulin. All these findings indicate a close association between a cross reactive antigen with the TASA of RSV-induced tumors and EGFR.


Assuntos
Antígenos de Neoplasias/análise , Antígenos de Superfície/análise , Carcinoma de Células Escamosas/imunologia , Receptores ErbB/metabolismo , Vírus do Sarcoma de Rous , Sarcoma Experimental/imunologia , Animais , Anticorpos Monoclonais , Carcinoma de Células Escamosas/metabolismo , Reações Cruzadas , Citotoxicidade Imunológica , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/fisiologia , Humanos , Insulina/farmacologia , Camundongos , Sarcoma Experimental/virologia , Células Tumorais Cultivadas/imunologia
12.
J Gen Virol ; 88(Pt 9): 2583-2589, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17698670

RESUMO

Walleye dermal sarcoma virus (WDSV) is a complex retrovirus associated with dermal sarcomas in walleye fish. Virus expression is tightly regulated and limited to accessory gene transcripts throughout tumour development. During tumour regression, this regulation is lost and the replication of virus is greatly enhanced. Cultured walleye fibroblasts infected in vitro do not produce significant quantities of infectious virus. Tissue culture cells established by explantation of tumour cells were found to harbour WDSV provirus and to express accessory and structural proteins. The sequence of the provirus showed little variation from a previous WDSV isolate. Retroviral particles were isolated from supernatants from these cells and were able to transfer infection to uninfected walleye fibroblasts. In addition to the virus present in supernatants, much of the virus was cell associated and liberated only by sonication. This virus was found at internal cellular membranes, including mitochondria, and was infectious.


Assuntos
Doenças dos Peixes/virologia , Sarcoma Experimental/virologia , Neoplasias Cutâneas/virologia , Pele/virologia , Animais , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , DNA Viral/genética , DNA Viral/isolamento & purificação , Doenças dos Peixes/patologia , Peixes , Dados de Sequência Molecular , Sarcoma Experimental/patologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/veterinária
14.
J Gen Virol ; 78 ( Pt 4): 893-903, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9129663

RESUMO

Small DNA tumour viruses, such as simian virus 40 (SV40), papilloma viruses and adenoviruses, encode proteins that form complexes with and inactivate the p53 and retinoblastoma (RB) proteins. This convergent evolution reflects the common need of these viruses to inactivate these two important regulators of cell cycle progression and cell survival. Polyomavirus, a close relative of SV40, is different. Its large T protein complexes only with RB, not with p53. We have examined whether this is compensated by the frequent appearance of p53 mutations in polyomavirus-induced tumours. We tested the p53 status of 15 polyomavirus-induced sarcomas. Two sarcomas were p53-negative while six carried mutant p53. Another six sarcomas expressed low levels of wild-type p53. One tumour expressed high levels of wild-type p53 protein as shown by DNA sequencing and immunofluorescence staining. MDM2 amplification was not detected in any of the tumours, but Northern blotting showed that MDM2 was overexpressed in at least two tumours that expressed wild-type p53 and in one tumour that expressed both wild-type and mutant p53. Treatment with the DNA-damaging agent mitomycin C caused p53 protein accumulation followed by induction of MDM2 and WAF1/p21 mRNA in four of the tumours expressing wild-type p53, indicating that p53-mediated transcriptional activation was unaltered in these tumours. However, p53-mediated transactivation of WAF1/p21 was impaired in the wild-type p53-expressing tumours that expressed elevated levels of MDM2. These results demonstrate that p53 mutation and inactivation are frequently but not invariably involved in polyomavirus-induced tumorigenesis.


Assuntos
Infecções por Polyomavirus/genética , Polyomavirus/isolamento & purificação , Sarcoma Experimental/genética , Proteína Supressora de Tumor p53/genética , Infecções Tumorais por Vírus/genética , Animais , Divisão Celular , Camundongos , Mutação Puntual , Sarcoma Experimental/patologia , Sarcoma Experimental/virologia , Transcrição Gênica
15.
Virology ; 237(1): 107-12, 1997 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-9344912

RESUMO

Walleye dermal sarcoma virus (WDSV) is a complex retrovirus associated with dermal sarcomas of walleye that develop and regress on a seasonal basis. WDSV contains, in addition to gag, pol, and env, three open reading frames (ORFs) designated ORF A, ORF B, and ORF C. The polymerase chain reaction technique was used to amplify and clone cDNAs representing subgenomic viral mRNAs isolated from developing (fall) and regressing (spring) tumors. Nine different singly or multiply spliced viral transcripts were identified and all were found to utilize a common 5' leader sequence. This leader sequence is spliced to the pol/env junction or downstream of env to generate singly spliced transcripts. Multiply spliced transcripts contain the 5' leader, the pol/env junction, and sequences derived from the 3' end of the genome. One multiply spliced transcript was isolated with the potential to encode the full-length ORF A protein. In addition, WDSV produced mRNAs that utilize alternative splice acceptor sites which would allow synthesis of five variant forms of the ORF A protein. In contrast, the ORF B protein is postulated to arise from a singly spliced transcript with the potential to encode the entire open reading frame. Spliced subgenomic transcripts representing ORF C mRNAs were not identified, suggesting that ORF C may be encoded from the full-length viral genomic transcript. We estimate that at least a 100-fold lower amount of the accessory/regulatory subgenomic transcripts exists in developing vs regressing tumors. These results demonstrate that WDSV undergoes an elaborate pattern of mRNA splicing similar to that of other complex retroviruses.


Assuntos
Peixes/virologia , Genes Virais , Genoma Viral , RNA Mensageiro/genética , RNA Viral/genética , Retroviridae/genética , Sarcoma Experimental/virologia , Neoplasias Cutâneas/virologia , Animais , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Viral/análise , Transcrição Gênica
16.
Virology ; 206(1): 85-92, 1995 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-7831844

RESUMO

hMt-c-fos-LTR transgenic mice (U. Rüther, D. Komitowski, F. R. Schubert, and E. F. Wagner. Oncogene 4, 861-865, 1989) developed bone sarcomas in 20% (3/15) of females at 448 +/- 25 days and in 8% (1/12) of males at 523 days. After infection of newborns with Akv, an infectious retrovirus derived from the ecotropic provirus of the AKR mouse, 69% (20/28) of female animals and 83% (24/29) of males developed malignant fibrous-osseous tumors. The tumors in infected transgenics developed with higher frequency and a 200-days shorter mean tumor latency period. The hMt-c-fos-LTR transgene was expressed in all the fibrous-osseous tumors. They also showed newly integrated Akv proviruses, but in most tumors Akv was detected and expressed in only a small number of the tumor cells. Wild-type C3H mice infected with Akv developed benign osteomas with an incidence of 33% and a latency period of 474 days. The data indicate that Akv exerts distinct pathogenic effects on the skeleton. In hMt-c-fos-LTR transgenic mice, predisposed to bone sarcomagenesis, Akv acts synergistically with the fos transgene, resulting in the development of fibrous-osseous tumors.


Assuntos
Vírus AKR da Leucemia Murina/patogenicidade , Neoplasias Ósseas/virologia , Genes fos , Sarcoma Experimental/virologia , Infecções Tumorais por Vírus/virologia , Animais , Sequência de Bases , Neoplasias Ósseas/genética , Primers do DNA , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Dados de Sequência Molecular , Sequências Repetitivas de Ácido Nucleico , Sarcoma Experimental/genética , Infecções Tumorais por Vírus/genética
17.
Scand J Immunol ; 45(1): 28-35, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9010497

RESUMO

A panel of sarcomas induced with 3-methylcholanthrene in normal and immunodeficient mice was studied for their capacity to present antigen by the endogenous, MHC class I restricted pathway. Lymphocytic choriomeningitis virus was used to infect cultured tumour cells, and the infected cells were tested for susceptibility to cytolysis by virus specific cytotoxic T cells. Tumour cells originating from tumours induced in immunocompetent C.B.-17 mice presented virus antigen more efficiently than tumour cells from immunodeficient SCID mice. No significant difference in virus antigen presentation was found between tumours from nude and nu/+ BALB/c mice. The sensitivity of target cells from the individual tumours to cytotoxic T lymphocyte (CTL) mediated lysis correlated negatively with their sensitivity to natural killer (NK) cell mediated lysis. There was a positive correlation between the sensitivity to CTL mediated lysis and surface expression of the MHC class I molecule Ld of the tumour cells. Tumour cells incapable of in vitro presentation of viral antigen to specific cytotoxic T cells originated from tumours known from previous experiments to be readily accepted after transplantation to immunocompetent, histocompatible recipients.


Assuntos
Citotoxicidade Imunológica/imunologia , Vigilância Imunológica/imunologia , Sarcoma Experimental/induzido quimicamente , Sarcoma Experimental/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Feminino , Antígenos H-2/biossíntese , Antígeno de Histocompatibilidade H-2D , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Vírus da Coriomeningite Linfocítica/isolamento & purificação , Metilcolantreno/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Sarcoma Experimental/metabolismo , Sarcoma Experimental/virologia
18.
Proc Natl Acad Sci U S A ; 98(2): 565-9, 2001 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-11209056

RESUMO

CpG islands are important in the protection of adjacent housekeeping genes from de novo DNA methylation and for keeping them in a transcriptionally active state. However, little is known about their capacity to protect heterologous genes and assure position-independent transcription of adjacent transgenes or retroviral vectors. To tackle this question, we have used the mouse aprt CpG island to flank a Rous sarcoma virus (RSV)-derived reporter vector and followed the transcriptional activity of integrated vectors. RSV is an avian retrovirus which does not replicate in mammalian cells because of several blocks at all levels of the replication cycle. Here we show that our RSV-derived reporter proviruses linked to the mouse aprt gene CpG island remain undermethylated and keep their transcriptional activity after stable transfection into both avian and nonpermissive mammalian cells. This effect is most likely caused by the protection from de novo methylation provided by the CpG island and not by enhancement of the promoter strength. Our results are consistent with previous finding of CpG islands in proximity to active but not inactive proviruses and support further investigation of the protection of the gene transfer vectors from DNA methylation.


Assuntos
Vírus do Sarcoma Aviário/genética , Ilhas de CpG , Vírus Defeituosos/genética , Regulação Viral da Expressão Gênica , Inativação Gênica , Vetores Genéticos/genética , Transcrição Gênica , Adenina Fosforribosiltransferase/genética , Animais , Vírus do Sarcoma Aviário/fisiologia , Linhagem Celular/virologia , Embrião de Galinha , Cricetinae , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Viral/química , DNA Viral/genética , Fibroblastos/virologia , Genes Reporter , Vetores Genéticos/fisiologia , Mesocricetus , Camundongos , Provírus/genética , Sarcoma Experimental/genética , Sarcoma Experimental/virologia , Sequências Repetidas Terminais , Integração Viral , Replicação Viral
19.
Acta Oncol ; 36(3): 323-30, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9208905

RESUMO

Radiation induced cytotoxicity was potentiated by neutralized metoclopramide (nMCA; Neu-Sensamide, Oxigene Inc) when a human lung adenocarcinoma (H2981) transplanted into scid mice and an adeno-type 12 virus induced mouse sarcoma (A12B3) inoculated into CBA mice were exposed in vivo to low dose radiation at single doses of 1 and 2 Gy respectively. However, when the radiation dose was increased to 6, 10 or 18 Gy (single dose) and combined with a single dose nMCA (2 mg/kg), tumor cytotoxicity was not sensitized by the combination treatment. A fractionated dose of ionizing radiation (3 x 1 Gy) in combination with nMCA at a repeated dose of 3 x 10 mg/kg body weight (1 dose/day, i.m.) significantly increased cytotoxicity in H2981 compared with radiation given alone. nMCA alone also had a statistically significant dose dependent cytotoxic effect on H2981 growth when it was administered as repeated doses (8 doses) at 2 mg/kg or 10 mg/kg (1 dose every second day), and a similar result was achieved at 20 mg/kg but not at 2 and 10 mg/kg in the A12B3 tumor. In addition, the tumor volume at the start of treatment was important for the anti-tumor effect of nMCA (i.e. the larger initial tumor volume gave less effect on tumor growth). Taken together, our data propose that the mode of action of nMCA is different from radiation, and hence the two mechanisms are at least additive when in combination with lower radiation doses. The data further suggest that the cytotoxic mechanism is consistent with potentiating apoptosis because low and repeated doses of radiation (1-2 Gy), which are known to increase cytotoxicity by apoptosis, are sensitized by nMCA but not high doses and nMCA has more potent anti-tumor effects against H2981 tumors which have a higher constitutive apoptotic fraction of cells than A12B3.


Assuntos
Metoclopramida/farmacologia , Radiossensibilizantes/uso terapêutico , Adenocarcinoma/patologia , Adenocarcinoma/radioterapia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Relação Dose-Resposta à Radiação , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Masculino , Metoclopramida/química , Camundongos , Camundongos Endogâmicos CBA , Camundongos SCID , Radiossensibilizantes/química , Dosagem Radioterapêutica , Sarcoma Experimental/patologia , Sarcoma Experimental/radioterapia , Sarcoma Experimental/virologia , Células Tumorais Cultivadas
20.
J Immunol ; 170(8): 4119-26, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12682242

RESUMO

Expression of adenovirus (Ad) serotype 2 or 5 (Ad2/5) E1A or human papillomavirus (HPV)16 E7 reportedly sensitizes cells to lysis by macrophages. Macrophages possess several mechanisms to kill tumor cells including TNF-alpha, NO, reactive oxygen intermediates (ROI), and Fas ligand (FasL). E1A sensitizes cells to apoptosis by TNF-alpha, and macrophages kill E1A-expressing cells, in part through the elaboration of TNF-alpha. However, E1A also up-regulates the expression of 70-kDa heat shock protein, a protein that inhibits killing by TNF-alpha and NO, thereby protecting cells from lysis by macrophages. Unlike E1A, E7 does not sensitize cells to killing by TNF-alpha, and the effector mechanism(s) used by macrophages to kill E7-expressing cells remain undefined. The purpose of this study was to further define the capacity of and the effector mechanisms used by macrophages to kill tumor cells that express Ad5 E1A or HPV16 E7. We found that Ad5 E1A, but not HPV16 E7, sensitized tumor cells to lysis by macrophages. Using macrophages derived from mice unable to make TNF-alpha, NO, ROI, or FasL, we determined that macrophages used NO, and to a lesser extent TNF-alpha, but not FasL or ROI, to kill E1A-expressing cells. Through the use of S-nitroso-N-acetylpenicillamine, which releases NO upon exposure to an aqueous environment, E1A was shown to directly sensitize tumor cells to NO-induced death. E1A sensitized tumor cells to lysis by macrophages despite up-regulating the expression of 70-kDa heat shock protein. In summary, E1A, but not E7, sensitized tumor cells to lysis by macrophages. Macrophages killed E1A-expressing cells through NO- and TNF-alpha-dependent mechanisms.


Assuntos
Proteínas E1A de Adenovirus/imunologia , Testes Imunológicos de Citotoxicidade/métodos , Proteínas de Choque Térmico HSP70/biossíntese , Macrófagos/imunologia , Óxido Nítrico/toxicidade , Proteínas Oncogênicas Virais/imunologia , Fator de Necrose Tumoral alfa/toxicidade , Regulação para Cima/imunologia , Células 3T3 , Proteínas E1A de Adenovirus/biossíntese , Adenovírus Humanos/imunologia , Animais , Linhagem Celular Transformada , Proteína Ligante Fas , Proteínas de Choque Térmico HSP70/fisiologia , Humanos , Imunização , Ligantes , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus , Sarcoma Experimental/imunologia , Sarcoma Experimental/prevenção & controle , Sarcoma Experimental/virologia , Superóxidos/metabolismo , Células Tumorais Cultivadas , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA