Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Physiol Mol Biol Plants ; 23(1): 197-205, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28250595

RESUMO

Chickpea (Cicer arietinum L.) is one of the most important legumes worldwide. We addressed this study to the genetic characterization of a germplasm collection from main chickpea growing countries. Several Italian traditional landraces at risk of genetic erosion were included in the analysis. Twenty-two simple sequence repeat (SSR) markers, widely used to explore genetic variation in plants, were selected and yielded 218 different alleles. Structure analysis and hierarchical clustering indicated that a model with three distinct subpopulations best fits the data. The composition of two subpopulations, named K1 and K2, broadly reflects the commercial classification of chickpea in the two types desi and kabuli, respectively. The third subpopulation (K3) is composed by both desi and kabuli genotypes. Italian accessions group both in K2 and K3. Interestingly, this study highlights genetic distance between desi genotypes cultivated in Asia and Ethiopia, which respectively represent the chickpea primary and the secondary centres of diversity. Moreover, European desi are closer to the Ethiopian gene pool. Overall, this study will be of importance for chickpea conservation genetics and breeding, which is limited by the poor characterization of germplasm collection.

2.
Br J Cancer ; 107(8): 1302-9, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-22929887

RESUMO

BACKGROUND: Human immune system (HIS)-engrafted mice are new tools to investigate human immune responses. Here, we used HIS mice to study human immune responses against human HER-2-positive cancer cells and their ability to control tumour growth and metastasis. METHODS: BALB/c Rag2(-/-), Il2rg(-/-) mice were engrafted with CD34(+) or CD133(+) human cord blood hematopoietic stem cells (HSC) and vaccinated with human HER-2-positive cancer cells SK-OV-3 combined to human IL-12. RESULTS: Both CD34(+) or CD133(+) human HSC gave long-term engraftment and differentiation, both in peripheral blood and in lymphoid organs, and production of human antibodies. Vaccinated mice produced specific anti-HER-2 human IgG. An s.c. SK-OV-3 challenge was significantly inhibited (but not abolished) in both vaccinated and non-vaccinated HIS mice. Tumours were heavily infiltrated with human and murine cells, mice showed NK cells and production of human interferon-γ, that could contribute to tumour growth inhibition. Vaccinated HIS mice showed significantly inhibited lung metastases when compared with non-vaccinated HIS mice and to non-HIS mice, along with higher levels of tumour-infiltrating human dendritic cells. CONCLUSION: Anti-HER-2 responses were elicited through an adjuvanted allogeneic cancer cell vaccine in HIS mice. Human immune responses elicited in HIS mice effectively inhibited lung metastases.


Assuntos
Antígenos CD34/imunologia , Antígenos CD/imunologia , Vacinas Anticâncer/imunologia , Glicoproteínas/imunologia , Neoplasias Pulmonares/imunologia , Peptídeos/imunologia , Receptor ErbB-2/imunologia , Antígeno AC133 , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células-Tronco Hematopoéticas/imunologia , Humanos , Isotipos de Imunoglobulinas/imunologia , Neoplasias Pulmonares/secundário , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C
3.
J Exp Med ; 194(9): 1195-205, 2001 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-11696586

RESUMO

Transgenic Balb/c mice expressing the transforming rat HER-2/neu oncogene develop early and multifocal mammary carcinomas. Within the first 5 months of life the tissue-specific expression of HER-2/neu causes a progression in all their 10 mammary glands from atypical hyperplasia to invasive carcinoma. It was previously observed that chronic administration of interleukin (IL)-12 increased tumor latency, but every mouse eventually succumbed to multiple carcinomas. A significant improvement in tumor prevention was sought by administering allogeneic mammary carcinoma cells expressing HER-2/neu combined with systemic IL-12. This treatment reduced tumor incidence by 90% and more than doubled mouse lifetime. For the maximum prevention p185(neu) antigen must be expressed by allogeneic cells. IL-12 treatment strongly increased the cell vaccine efficacy. The mammary glands of mice receiving the combined treatment displayed a markedly reduced epithelial cell proliferation, angiogenesis, and HER-2/neu expression, while the few hyperplastic foci were heavily infiltrated by granulocytes, macrophages, and CD8(+) lymphocytes. Specific anti-HER-2/neu antibodies were produced and a nonpolarized activation of CD4(+) and CD8(+) cells secreting IL-4 and interferon (IFN)-gamma were evident. A central role for IFN-gamma in the preventive effect was proven by the lack of efficacy of vaccination in IFN-gamma gene knockout HER-2/neu transgenic Balb/c mice. A possible requirement for IFN-gamma is related to its effect on antibody production, in particular on IgG2a and IgG2b subclasses, that were not induced in IFN-gamma knockout HER-2/neu mice. In conclusion, our data show that an allogeneic HER-2/neu-expressing cell vaccine combined with IL-12 systemic treatment can prevent the onset of genetically determined tumors.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Interleucina-12/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/fisiologia , Animais , Mama/patologia , Linfócitos T CD4-Positivos/imunologia , Transplante de Células , Feminino , Imunidade Celular/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/administração & dosagem , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Ratos , Receptor ErbB-2/genética , Transplante Homólogo , Células Tumorais Cultivadas , Vacinação/métodos
4.
J Exp Med ; 188(3): 589-96, 1998 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-9687535

RESUMO

The ability of interleukin (IL)-12 to prevent tumors when administered to individuals with a genetic risk of cancer was studied in two lines of transgenic mice expressing rat HER-2/neu oncogene in the mammary gland. Female BALB/c (H-2(d)) mice carrying the activated HER-2/ neu oncogene show no morphological abnormalities of the mammary gland until 3 wk of age. They then progress through atypical hyperplasia to in situ lobular carcinoma and at 33 wk of age all 10 mammary glands display invasive carcinomas. Adult FVB mice (H-2(q)) carrying the HER-2/neu protooncogene develop mammary carcinomas with a longer latency (38-49 wk) and a lower multiplicity (mean of 2.6 tumors/mice). Treatment with IL-12 (5 daily intraperitoneal injections, 1 wk on, 3 wk off; the first course with 50 ng IL-12/day, the second with 100 ng IL-12/day) begun at 2 wk of age in BALB/c mice and at 21 wk of age in FVB mice markedly delayed tumor onset and reduced tumor multiplicity. Analogous results were obtained in immunocompetent and permanently CD8(+) T lymphocyte-depleted mice. In both transgenic lines, tumor inhibition was associated with mammary infiltration of reactive cells, production of cytokines and inducible nitric oxide synthase, and reduction in microvessel number, in combination with a high degree of hemorrhagic necrosis.


Assuntos
Antineoplásicos/farmacologia , Carcinoma in Situ/prevenção & controle , Carcinoma Lobular/prevenção & controle , Interleucina-12/farmacologia , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/fisiologia , Animais , Antineoplásicos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma in Situ/genética , Carcinoma in Situ/imunologia , Carcinoma Lobular/genética , Carcinoma Lobular/imunologia , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Feminino , Interferon gama/imunologia , Interleucina-12/imunologia , Depleção Linfocítica , Masculino , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Ratos , Receptor ErbB-2/genética , Fator de Necrose Tumoral alfa/imunologia , Molécula 1 de Adesão de Célula Vascular/biossíntese
5.
J Cell Biol ; 151(3): 495-506, 2000 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-11062252

RESUMO

DRAL is a four and a half LIM domain protein identified because of its differential expression between normal human myoblasts and the malignant counterparts, rhabdomyosarcoma cells. In the current study, we demonstrate that transcription of the DRAL gene can be stimulated by p53, since transient expression of functional p53 in rhabdomyosarcoma cells as well as stimulation of endogenous p53 by ionizing radiation in wild-type cells enhances DRAL mRNA levels. In support of these observations, five potential p53 target sites could be identified in the promoter region of the human DRAL gene. To obtain insight into the possible functions of DRAL, ectopic expression experiments were performed. Interestingly, DRAL expression efficiently triggered apoptosis in three cell lines of different origin to the extent that no cells could be generated that stably overexpressed this protein. However, transient transfection experiments as well as immunofluorescence staining of the endogenous protein allowed for the localization of DRAL in different cellular compartments, namely cytoplasm, nucleus, focal contacts, as well as Z-discs and to a lesser extent the M-bands in cardiac myofibrils. These data suggest that downregulation of DRAL might be involved in tumor development. Furthermore, DRAL expression might be important for heart function.


Assuntos
Apoptose , Proteínas de Homeodomínio , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição , Ativação Transcricional , Proteína Supressora de Tumor p53/fisiologia , Animais , Sequência de Bases , Caspases/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Clonagem Molecular , Citoplasma/metabolismo , Imunofluorescência , Raios gama , Perfilação da Expressão Gênica , Humanos , Proteínas com Homeodomínio LIM , Camundongos , Proteínas Musculares/genética , Miocárdio/citologia , Miocárdio/metabolismo , Miofibrilas/metabolismo , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética , Estrutura Terciária de Proteína , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Elementos de Resposta/genética , Rabdomiossarcoma/genética , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Ativação Transcricional/efeitos da radiação , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/efeitos da radiação
6.
J Nutr Biochem ; 69: 1-9, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31035100

RESUMO

Folate-dependent one-carbon cycle metabolism (FOCM) plays a critical role in maintaining genomic stability through regulating DNA biosynthesis, repair and methylation. Folate metabolites as well as other metabolites in the FOCM are hypothesized to be altered when cells transition from normal to cancerous state. Using cells at different stages in their development into colorectal cancer, the FOCM metabolites were profiled as an effort to phenotype the cells, and the metabolite levels were compared to the expressions of related genes. Here, we investigate whether there is a correlation between the metabolite levels, DNA methylation levels and the expression of the related genes that drive the levels of these metabolites. Using CRL1459, APC10.1, HCT116 and Caco-2, we show for the first time that FOCM metabolites correlate with the gene expression patterns. These differences follow a trend that may facilitate distinguishing colon cells at the different stages as they transition into cancerous state. The folate distribution and methionine levels were found to be key in determining the staging of the colon cells in CRC development. Also, expression of CBS, MTRR and MAT genes may facilitate distinguishing between untransformed and transformed colon cells.


Assuntos
Carbono/metabolismo , Colo/patologia , Ácido Fólico/metabolismo , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transformação Celular Neoplásica , Colo/citologia , Colo/metabolismo , Metilação de DNA , Ferredoxina-NADP Redutase/genética , Receptor 1 de Folato/genética , Ácido Fólico/genética , Ácido Fólico/farmacologia , Regulação da Expressão Gênica , Glicina Hidroximetiltransferase/genética , Células HCT116 , Humanos
7.
Curr Cancer Drug Targets ; 5(3): 221-8, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15892621

RESUMO

Prevention of cancer through the activation of the immune system has been explored in recent years in preclinical systems thanks to the availability of several new transgenic mouse models that closely mimic the natural history of human tumors. The most thoroughly investigated model of cancer immunoprevention is the mammary carcinoma of HER-2/neu transgenic mouse. In this system it has clearly been shown that the activation of immune defences in healthy individuals can effectively prevent the subsequent onset of highly aggressive mammary carcinomas. A complete prevention was obtained using a combination of three signals (the so called "triplex" vaccine) that included the specific antigen (p185, the product of HER-2/neu) and nonspecific signals like allogeneic histocompatibility antigens and interleukin 12. The analysis of protective immune responses in models of cancer immunoprevention revealed some unexpected features, in particular the central role of antibodies in immunoprevention, at variance with conventional immuno-therapy which is firmly based on cytotoxic T cells. In the HER-2/neu system anti-p185 antibodies, in addition to immunological functions leading to tumor cell lysis, inhibit p185 dimerization and induce its internalization, resulting in the inhibition of mitogenic signaling. Most current tumor antigens appear to be unsuitable targets for cancer immunoprevention. An ideal antigen should have a crucial pathogenetic role in tumor growth to avoid the selection of antigen loss variants. Downregulation of major histocompatibility complex (MHC) expression during tumor progression frequently limits antigen recognition by MHC-restricted T cells. Thus an ideal antigen for cancer immunoprevention should be recognized both by T cells and by antibodies. Antibody binding to cell surface oncogenic determinants, in addition to complement- and cell-mediated tumor cell lysis, can block mitogenic signaling and induce internalization, resulting in tumor growth arrest. A search for new tumor antigens should be conducted among molecules that are directly involved in neoplastic transformation and are recognizable by the immune response also in MHC loss variants. Novel tumor antigens fulfilling both conditions will be crucial for the development of cancer immunoprevention and will provide new targets also for cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Neoplasias/prevenção & controle , Animais , Humanos , Camundongos , Camundongos Transgênicos
8.
J Natl Cancer Inst ; 76(1): 87-93, 1986 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3455746

RESUMO

Cells from TS/A, a metastasizing line derived from a spontaneous BALB/c mouse mammary adenocarcinoma, were injected either sc or iv in syngeneic mice, and the resulting lung metastases or lung colonies were briefly cultured in vitro and reinjected in mice by the same route; this procedure was repeated 10 times. All the variants obtained did not show a metastatic capacity higher than the parental cell line. Moreover, they gave a number of metastases significantly lower than that produced by high metastatic clones selected in vitro from TS/A. The number of lung colonies obtained with intravenously selected (COL) variants was significantly higher than that obtained with subcutaneously selected (META) variants, TS/A, or in vitro-selected clones; this was already observable after the first cycle of selection. Both COL and META variants did not show the in vitro growth properties of the in vitro-selected high metastatic clones. In conclusion, both intravenous and subcutaneous selection procedures did not lead to an enrichment in high metastatic populations, even if such populations were present in the parental line and had been cloned in vitro; only the intravenous procedure selected high-colonizing variants.


Assuntos
Metástase Neoplásica/patologia , Animais , Linhagem Celular , Células Clonais , Feminino , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C
9.
Cancer Res ; 49(13): 3631-6, 1989 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-2471586

RESUMO

The effect of various antineoplastic drugs (1-beta-D-arabinofuranosylcytosine, 5-azacytidine, cisplatin, dactinomycin, epirubicin, vincristine, and the activated metabolite of cyclophosphamide, mafosfamide) on cell differentiation in vitro was investigated using a human alveolar rhabdomyosarcoma cell clone, RMZ-RC2. These cells are able to differentiate spontaneously from small mononuclear proliferating elements to terminal, extremely elongated multinuclear structures resembling myotubes; morphological differentiation is accompanied by the expression of myosins, in particular the embryonic isoform, which was used in this study as a specific marker of myogenic differentiation. The proportion of differentiated myosin-positive cells, which was around 10-15% in control cultures 10-15 days after seeding, was increased by some drug treatments up to 30-40%; the proportion of multinuclear elements was also increased. 1-beta-D-arabinofuranosylcytosine and 5-azacytidine were the most effective drugs, while dactinomycin had no effect; other molecules ranked in between. Since significant increments were usually observed after treatment with drug doses inhibiting cell growth, the kinetic behavior of the absolute number of myosin-positive cells or nuclei was analyzed to assess whether some effects could be due to a negative selection of proliferating, undifferentiated cells. This appeared to be the case for vincristine and epirubicin, while 1-beta-D-arabinofuranosylcytosine, 5-azacytidine, and, to a lesser degree, mafosfamide and cisplatin actually seemed to increase differentiation ability.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Músculos/citologia , Rabdomiossarcoma/patologia , Azacitidina/farmacologia , Divisão Celular/efeitos dos fármacos , Fusão Celular , Cisplatino/farmacologia , Ciclofosfamida/farmacologia , Citarabina/farmacologia , Humanos , Técnicas In Vitro , Miosinas/metabolismo , Células Tumorais Cultivadas
10.
Cancer Res ; 56(19): 4503-8, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8813148

RESUMO

Carcinoembryonic antigen (CEA) is a highly glycosylated cell surface glycoprotein belonging to the immunoglobulin superfamily. CEA has been involved in vitro in adhesion mechanisms, but little is known about the function of this glycoprotein in vivo in normal tissue differentiation and malignancy. With regard to the relationship between CEA expression and tissue differentiation, it has been reported that transfection of the CEA gene in rat L6 myoblasts results in a complete block of myogenic differentiation. To extend investigations to the transformed myogenic counterpart and examine CEA effects on differentiation and malignancy outside the colon system, we have transfected the human CEA gene in human rhabdomyosarcoma cells. Human rhabdomyosarcoma cells transfected with the CEA gene correctly expressed membrane CEA anchored via glycosylphosphatidylinositol and secreted CEA in the medium. CEA gene transfer in human rhabdomyosarcoma cells, which display a limited differentiation ability, does not further inhibit myogenic differentiation or alter in vitro proliferation or natural killer sensitivity. CEA transfection does not affect s.c. growth in nude mice, but the ectopic expression of CEA in human rhabdomyosarcoma cells can strongly inhibit their metastatic ability to lungs and adrenals after i.v. injection. The impairment of metastatic potential correlates with a reduction in the homotypic adhesion properties of the cells. These data suggest that CEA, in some systems, can interfere with intercellular adhesion and, at least for cells not metastatic to the liver, can act as an anti-metastatic molecule.


Assuntos
Antígeno Carcinoembrionário/fisiologia , Regulação Neoplásica da Expressão Gênica , Metástase Neoplásica/genética , Rabdomiossarcoma/genética , Neoplasias de Tecidos Moles/genética , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/secundário , Animais , Antígeno Carcinoembrionário/biossíntese , Antígeno Carcinoembrionário/genética , Diferenciação Celular , DNA Complementar/genética , Feminino , Glicosilfosfatidilinositóis/metabolismo , Humanos , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Músculos/patologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Rabdomiossarcoma/patologia , Rabdomiossarcoma/secundário , Neoplasias de Tecidos Moles/patologia , Transfecção , Células Tumorais Cultivadas
11.
Cancer Res ; 58(6): 1225-30, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9515809

RESUMO

Immunological gene therapy of cancer relies heavily on the activation of T cells, but tumors with defects in MHC gene expression are not recognized by MHC-restricted T cells. To investigate the potential of cytokine genes for the therapy of MHC-negative tumors, we transduced B78H1, a class I-negative murine melanoma clone, with a polycistronic vector carrying murine interleukin (IL)-12 genes. The clones studied produced 400-25,000 pg/ml IL-12; their in vitro growth properties were similar to those of parental cells. A complete inhibition of growth was observed in vivo both after s.c. and i.v. administration of all IL-12 clones. IL-12-transduced cells were also used as a therapeutic vaccine in mice bearing micrometastases by nontransduced parental cells. A significant (80-90%) reduction in the number of lung nodules was obtained. Immunohistochemical analysis and studies in immunocompromised hosts showed that T cells and natural killer cells had a significant role in the elimination of IL-12-releasing cells. In situ hybridization with cytokine probes detected a strong increase in the proportion of leukocytes positive for IFN-gamma, tumor necrosis factor alpha, IL-1beta, and IFN-inducible protein 10 at the site of rejection of IL-12-engineered tumor cells. However, it was clear that the loss of in vivo growth was also due to T-cell- and natural killer cell-independent factors, possibly related to the antiangiogenic properties of IL-12. In conclusion, tumor therapy based on IL-12 gene transduction was effective on a MHC-negative metastatic tumor, suggesting a possible application to MHC-defective human neoplasms.


Assuntos
Antígenos H-2 , Interleucina-12/genética , Complexo Principal de Histocompatibilidade , Melanoma Experimental/terapia , Metástase Neoplásica/prevenção & controle , Animais , Vacinas Anticâncer/imunologia , Citotoxicidade Imunológica , Terapia Genética , Hospedeiro Imunocomprometido , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neovascularização Patológica , Transdução Genética
12.
Cancer Res ; 56(17): 3898-901, 1996 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-8752155

RESUMO

Human rhabdomyosarcoma cells express membrane epidermal growth factor receptor (ECF-R), which could confer responsiveness to EGF and transforming growth factor-alpha (TGF-alpha) of autocrine or paracrine origin. To study the role played by this growth factor circuit in the proliferation and differentiation of myogenic neoplastic cells, human rhabdomyosarcoma EGF-R-expressing cells (RD/18 clone) have been transfected with a plasmid containing a fragment of the EGF-R cDNA in the antisense orientation. In vitro growth and differentiative ability were studied on six antisense-transfected clones (AS) in comparison to parental RD/18 cells and to cells transfected with the plasmid containing only the neomycin resistance gene (NEO). A reduced EGF-R membrane expression was found in AS clones by decreased immunofluorescence with an anti-EGF-R monoclonal antibody. All AS transfectants had a greatly impaired proliferative ability, even when cultured in fetal bovine serum-containing medium. Proliferation of AS clones was completely blocked in medium supplemented with 2% horse serum. The differentiation ability of AS clones was heterogeneous, ranging from clones with a percentage of myosin-positive cells higher than controls to clones with a negligible myosin expression. Therefore, the growth impairment determined by the loop interruption is not sufficient to switch on the differentiation program. The role played by EGF-R in the proliferation of human rhabdomyosarcoma cells suggests that this receptor could constitute a target for a therapeutic approach.


Assuntos
DNA Complementar/genética , Receptores ErbB/fisiologia , Oligonucleotídeos Antissenso/genética , Rabdomiossarcoma/patologia , Animais , Sequência de Bases , Bovinos , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Receptores ErbB/biossíntese , Receptores ErbB/genética , Humanos , Dados de Sequência Molecular , Rabdomiossarcoma/genética , Rabdomiossarcoma/metabolismo , Transfecção , Células Tumorais Cultivadas
13.
Oncogene ; 18(3): 739-46, 1999 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-9989824

RESUMO

The relationship between P-glycoprotein expression and malignancy is controversial. We have recently found that, in osteosarcoma, multidrug resistance (MDR) is associated with a less aggressive behavior, both in vitro and in clinical settings. In this study, we evaluated whether P-glycoprotein overexpression has a cause-effect relationship with the reduced metastatic potential of MDR cells, or rather reflects a more complex phenotype. MDR1 gene-transfected osteosarcoma cell clones, showing different levels of P-glycoprotein expression, were analysed for their in vitro characteristics and their tumorigenic and metastatic ability in athymic mice. Apart from the different levels of P-glycoprotein, no significant change in the expression of surface antigens or in the differentiative features were observed in the MDR1 gene transfectants compared to the parental cell lines or control clones, obtained by transfection with neo gene alone. In contrast to controls, however, MDR1 transfectants showed a significantly lower ability to grow in semi-solid medium and were completely unable to grow and give lung metastases in athymic mice. These findings indicate that P-glycoprotein overexpression is causally associated with a low malignant potential of osteosarcoma cells, and open new insights on the role and functions of P-glycoprotein activity.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Osteossarcoma/fisiopatologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Humanos , Camundongos , Camundongos Nus , Fenótipo , Transfecção , Células Tumorais Cultivadas
14.
Oncogene ; 12(8): 1697-705, 1996 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-8622890

RESUMO

The MET oncogene encodes the receptor for HGF/Scatter Factor, known to control cell motility and invasion in epithelial cells. We report that the Met/HGF receptor, absent in differentiated adult skeletal muscles, is aberrantly expressed in clinical samples and in established cell lines of human rhadbomyosarcomas. In both the embryonal and alveolar histotypes the oncogene is overexpressed and, in some cases, amplified. The Met receptor is exposed at the cell surface and is functionally active in response to HGF/Scatter Factor. Accordingly, rhabdomyosarcoma cells exhibit an invasive phenotype in vitro in response to exogenous HGF/Scatter factor. As the factor is known to be produced by connective tissues, a paracrine stimulation of rhabdomyosarcoma invasiveness in vivo is hypothesized. Two alveolar rhabdomyosarcomas were found in co-express the ¿two-kringle' alternatively-spliced HGF/Scatter Factor variant, which has been previously shown to stimulate cell motility and matrix invasion in vitro. These cells displayed the invasive phenotype in the absence of exogenous HGF/Scatter Factor, suggesting an autocrine mechanism in vivo. These data indicate that aberrant expression of the MET proto-oncogene provides rhabdomyosarcoma cells with the same property as embryonal myoblasts to migrate into the surrounding connective tissues.


Assuntos
Invasividade Neoplásica/genética , Receptores Proteína Tirosina Quinases/genética , Rabdomiossarcoma Alveolar/genética , Rabdomiossarcoma Alveolar/patologia , Rabdomiossarcoma Embrionário/genética , Rabdomiossarcoma Embrionário/patologia , Sequência de Bases , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Proteínas Tirosina Quinases/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-met , Células Tumorais Cultivadas
15.
Int J Immunopathol Pharmacol ; 18(2): 351-63, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15888257

RESUMO

Prevention of the progression of precancerous lesions by vaccines is virtually uncharted territory. Their potential, however, is being assessed in transgenic mice which develop autochthonous tumors with defined stages of progression. In this paper we show that the DNA micro-array technology significantly helps assessment of the preventive efficacy of a combined DNA and cell vaccine. All female rat Her-2/neu transgenic BALB/c (BALB-neuT) mice develop an invasive carcinoma in each of their mammary glands within 25 weeks of age. This is elicited by the activated transforming rat Her-2/neu oncogene embedded in their genome. We have previously shown that vaccination of mice bearing multiple in situ carcinomas with DNA plasmids which code for the extracellular and transmembrane domain of rat p185neu, the product of the rat Her-2/neu oncogene, followed by a boost with rat p185neu+ allogeneic cells engineered to secrete interferon-gamma, keeps 48% of mice tumor free until week 32. We have now extended our follow-up until mice reach one year of age and show that protection vanishes as time progresses. This observation suggests that the accuracy of the results studying immunotherapy against life-threatening tumors is a function of the length of the follow-up. The application of microarrays, and the concordance of morphologic and gene expression data led us to identify antibody as the main mechanism induced by vaccination. Protection is associated with a break of tolerance and a limited autoimmunity against the endogenous mouse p185neu.


Assuntos
Autoimunidade/efeitos dos fármacos , Glicoproteínas/genética , Neoplasias Mamárias Experimentais/prevenção & controle , Lesões Pré-Cancerosas/terapia , Receptor ErbB-2/genética , Vacinas de DNA/uso terapêutico , Animais , Linhagem Celular Tumoral , Clonagem Molecular , Feminino , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Lesões Pré-Cancerosas/imunologia , Lesões Pré-Cancerosas/patologia , Ratos , Transgenes
16.
Clin Cancer Res ; 4(8): 1843-9, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9717810

RESUMO

CD40, a membrane glycoprotein of the tumor necrosis factor receptor family, is expressed by several tumor types, including B-cell lymphomas, carcinomas, and melanoma, but little is known concerning its expression by sarcoma. We used flow cytometry to analyze the expression of CD40 in human cell lines derived from 12 osteosarcomas, 6 Ewing's sarcomas, and 5 rhabdomyosarcomas. Detectable CD40 levels ranging from low to very high were found in one-third of osteosarcomas, whereas five of six Ewing's sarcomas expressed intermediate levels of CD40; all rhabdomyosarcomas were CD40-negative. At the tissue level, two of eight primary high-grade osteosarcomas showed CD40-positive immunostaining. Osteosarcoma cells and Ewing's sarcoma cells expressing CD40 were treated with recombinant soluble CD40 ligand to analyze CD40 function. Treatment with soluble CD40 ligand increased the level of apoptotic cells and stimulated the transcription of matrix metalloproteinase 9 gene, enhancing matrix metalloproteinase 9 enzyme secretion. The results indicate that in human osteosarcoma and Ewing's sarcoma, CD40 is a functional receptor whose engagement can have opposite effects on tumor cell survival and malignancy.


Assuntos
Neoplasias Ósseas/metabolismo , Antígenos CD40/biossíntese , Osteossarcoma/metabolismo , Sarcoma de Ewing/metabolismo , Apoptose/efeitos dos fármacos , Biópsia , Neoplasias Ósseas/enzimologia , Neoplasias Ósseas/patologia , Ligante de CD40 , Colagenases/biossíntese , Colagenases/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Metaloproteinase 9 da Matriz , Glicoproteínas de Membrana/farmacologia , Osteossarcoma/enzimologia , Osteossarcoma/patologia , Rabdomiossarcoma/enzimologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Sarcoma de Ewing/enzimologia , Sarcoma de Ewing/patologia , Células Tumorais Cultivadas
17.
Hum Gene Ther ; 6(6): 743-52, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7548274

RESUMO

The mouse mammary carcinoma TS/A, of BALB/c (H-2d) origin, was transfected with the murine interferon-gamma (IFN-gamma) gene (Int. J. Cancer 55: 320, 1993). We used IFN-gamma transfectants as recipients for a second round of transfections with murine allogeneic class I histocompatibility (H-2b) genes that are modulated by IFN. Transfectants with either gene alone, as well as parent TS/A cells (TS/A-pc), were used as controls. Only double transfectants expressed high levels of the allogeneic H-2b genes, while in H-2b single transfectants the expression was very low (but was induced by treatment with exogenous IFN-gamma). The tumorigenic potential of IFN-gamma or H-2b single transfectants was reduced in comparison to TS/A-pc. IFN-gamma+H-2Kb double transfectants were almost nontumorigenic, while IFN-gamma+H-2Db clones gave rise to tumors in about one-half of mice. The experimental metastatic ability of all IFN-gamma+H-2b double transfectants was very low. IFN-gamma single transfectants were known to induce a strong macrophage response in the host. The expression of allogeneic H-2 antigens added a T-lymphocyte-mediated response that accounted for the lower tumorigenicity of double transfectants. These results show that it is possible to steer the immune response evoked by tumor cells for therapeutic purposes. Moreover, the high H-2 expression obtained in IFN-gamma+H-2b double transfectants suggests that single IFN-gamma transfectants are ideal recipients for all IFN-sensitive genes. This approach can be used also for other general-purpose inducers of gene expression.


Assuntos
Antígenos H-2/genética , Interferon gama/genética , Neoplasias Mamárias Experimentais/genética , Metástase Neoplásica/genética , Transdução Genética , Animais , Citotoxicidade Imunológica , Feminino , Hospedeiro Imunocomprometido , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica/prevenção & controle , Transfecção , Células Tumorais Cultivadas
18.
Hum Gene Ther ; 9(2): 217-24, 1998 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9472781

RESUMO

The parental cells of the TSA murine mammary adenocarcinoma (TSA-pc) were transfected with both the interferon-gamma (IFN-y) gene and the cytosine deaminase (CD) suicide gene to obtain a therapeutic vaccine active against TSA-pc lung metastases. Even in the absence of treatment with the prodrug 5-fluorocytosine (5-FC), the local growth of double transfectants (CD-y clones) was inhibited by a marked recruitment of granulocytes and macrophages. In mice harboring TSA-pc micrometastases, therapeutic vaccination with either IFN-gamma or CD single transfectants reduced the number of lung nodules, whereas CD-gamma double transfectants abrogated metastasis growth in up to 80% of mice. Treatment of mice with 5-FC did not alter the curative efficacy of CD-gamma double-transfectant cells. By contrast, in mice vaccinated with CD single-transfectant cells, 5-FC treatment caused a significant loss of their curative activity. Host T cells played an active role in the cure of lung metastases, because vaccination of nude mice with CD-gamma cells was uneffective.


Assuntos
Adenocarcinoma/genética , Interferon gama/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Neoplasias Mamárias Experimentais/genética , Nucleosídeo Desaminases/genética , Transfecção , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Citosina Desaminase , Citotoxicidade Imunológica , Feminino , Terapia Genética , Imunoterapia Ativa , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Linfócitos T Citotóxicos/imunologia
19.
Gene ; 274(1-2): 139-49, 2001 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-11675006

RESUMO

Rhabdomyosarcoma is a soft tissue tumor committed to the myogenic lineage but arrested prior to terminal differentiation. To identify new genes implicated in the block in myogenic differentiation of rhabdomyosarcoma cells and those responsible for their proceeding along the myogenic pathway we used cDNA microarrays to compare gene expression profiles of two clones of the human embryonal rhabdomyosarcoma cell line RD with different myogenic potentials: RD/12, which is unable to differentiate, and RD/18, which shows elements able to terminally differentiate, as defined by the expression of myosin heavy chain (up to 50% of the population) and the formation of multinucleated myotube-like structures. We identified 80 genes differentially expressed by the two clones. Differentiating RD/18 cells overexpressed the myogenic transcription factor myogenin along with known myogenic markers; myogenin transfection into undifferentiated RD/12 cells was able to revert the phenotype giving rise to 94% of clones displaying a differentiated morphology. RD/18 cells also expressed several genes not known to be expressed in rhabdomyosarcoma or muscle cells, such as pigment-epithelium derived factor and endothelin-3. Poorly differentiated RD/12 cells, along with genes related to mesenchymal lineage or early myogenic commitment, also expressed genes not previously known to be related to the differentiation block of human rhabdomyosarcoma, such as monocyte chemotactic protein-1, connective tissue growth factor and insulin-like growth factor binding protein-5. Differential expression of these genes in a time course of differentiation suggested their potential roles as either new myogenic markers or repressors of differentiation. These results identify a cluster of new genes related to the aberrant myogenic differentiation program of human rhabdomyosarcoma cells.


Assuntos
Diferenciação Celular/genética , Perfilação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular , Músculos/metabolismo , Rabdomiossarcoma/genética , Quimiocina CCL2/genética , Fator de Crescimento do Tecido Conjuntivo , Relação Dose-Resposta a Droga , Endotelina-3/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Substâncias de Crescimento/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Músculos/citologia , Miogenina/genética , Miosinas/genética , Análise de Sequência com Séries de Oligonucleotídeos , Rabdomiossarcoma/patologia , Fatores de Tempo , Células Tumorais Cultivadas
20.
Clin Exp Metastasis ; 4(1): 13-23, 1986.
Artigo em Inglês | MEDLINE | ID: mdl-3698365

RESUMO

The expression of steroid receptors and the in vitro responsiveness to steroids were used to investigate the cell heterogeneity of a BALB/c mammary carcinoma cell line (TS/A) by means of its high- and low-metastatic clones previously selected in vitro. All the clones studied contained appreciable levels of receptors for oestrogens and for glucocorticoids. The in vitro responses of clones to 17 beta-oestradiol were very poor and comparable; conversely, a heterogeneous pattern of responsiveness to glucocorticoids was observed. In the presence of dexamethasone, the in vitro growth of high-metastatic clones was either unaffected or stimulated and dome formation was significantly increased. Dexamethasone treatment of low-metastatic clones caused inhibition of in vitro proliferation and a morphological shift from a fibroblast-like growth pattern towards the epithelial phenotype. One out of the three low-metastatic clones tested acquired the ability to form domes in the presence of dexamethasone, albeit sporadically. The in vitro treatment with dexamethasone significantly increased the lung colonization ability of the two low-metastatic clones studied, whereas no significant effect was observed with high-metastatic clones. Data presented here suggest that TS/A cell line consists of heterogeneous populations with peculiar proliferative and differentiative responses to glucocorticoids.


Assuntos
Dexametasona/farmacologia , Neoplasias Mamárias Experimentais/patologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Células Clonais/patologia , Estrogênios/farmacologia , Etanol/farmacologia , Feminino , Neoplasias Pulmonares/secundário , Camundongos , Metástase Neoplásica , Receptores de Esteroides/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA