Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Biol ; 97(4): 1191-9, 1983 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-6194162

RESUMO

Two proteins of 53,000 and 56,000 mol wt have been found to be associated with coated vesicles (CV) purified from bovine brain and chicken liver. These proteins share molecular weights, isoelectric points, and antigenic determinants with alpha- and beta-tubulins purified from bovine brain. Based on SDS PAGE and electron microscopic analysis of controlled pore glass bead exclusion column fractions, both the tubulins and the major CV polypeptide clathrin were found to chromatograph as components of a single kinetic particle. In addition, tubulin and CV antigens assayed by a sensitive enzyme-linked-immunoadsorbent method eluted from the columns with constant stoichiometry. These data provide evidence that tubulin is a molecular component of coated vesicles.


Assuntos
Endossomos/análise , Tubulina (Proteína)/análise , Animais , Bovinos , Galinhas , Clatrina/análise , Ensaio de Imunoadsorção Enzimática , Epitopos , Ponto Isoelétrico , Peso Molecular , Tubulina (Proteína)/imunologia
2.
J Clin Invest ; 92(6): 3017-22, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8254056

RESUMO

The signal transduction mechanisms involved in tumor cell adhesion to endothelial cells are still largely undefined. The effect of metastatic murine melanoma cell and human prostate carcinoma cell contact on cytosolic [Ca2+] of bovine artery endothelial cells was examined in indo-1-loaded endothelial cell monolayers. A rapid increase in endothelial cell [Ca2+] occurred on contact with tumor cells, but not on contact with 8-microns inert beads. A similar increase in endothelial cell [Ca2+] was observed with human neutrophils or monocyte-like lymphoma cells, but not with endothelial cells, red blood cells, and melanoma cell-conditioned medium. The increase in endothelial cell [Ca2+] was not inhibited by extracellular Ca2+ removal. In contrast, endothelial cell pretreatment with thapsigargin, which releases endoplasmic reticulum Ca2+ into the cytosol and depletes this Ca2+ store site, abolished the cytosolic [Ca2+] rise upon melanoma cell contact. Endothelial cell pretreatment with the membrane-permeant form of the Ca2+ chelator bis-(O-aminophenoxyl)ethane-N,N,N',N'-tetraacetic acid blocked the increase in cytosolic [Ca2+]. Under static and dynamic flow conditions (0.46 dyn/cm2) bis-(O-aminophenoxyl)ethane-N,N,N',N'-tetraacetic acid pretreatment of bovine pulmonary artery endothelial cell monolayers inhibited melanoma cell adhesion to the endothelial cells. Thus, tumor cell contact with endothelial cells induces a rapid Ca2+ release from endothelial intracellular stores, which has a functional role in enhancing cell-cell adhesion.


Assuntos
Cálcio/fisiologia , Adesão Celular , Endotélio Vascular/fisiologia , Melanoma Experimental/fisiopatologia , Neoplasias da Próstata/fisiopatologia , Animais , Cálcio/metabolismo , Bovinos , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Ácido Egtázico/farmacologia , Endotélio Vascular/metabolismo , Eritrócitos/fisiologia , Corantes Fluorescentes , Humanos , Indóis , Cinética , Masculino , Camundongos , Artéria Pulmonar , Transdução de Sinais , Células Tumorais Cultivadas
3.
Nat Biotechnol ; 19(3): 219-24, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11231553

RESUMO

Previously we showed the superior in vitro survival of human telomerase reverse transcriptase (hTERT)-transduced human endothelial cells (EC). Here we show that retroviral-mediated transduction of hTERT in human dermal microvascular EC (HDMEC) results in cell lines that form microvascular structures when subcutaneously implanted in severe combined immunodeficiency (SCID) mice. Anti-human type IV collagen basement membrane immunoreactivity and visualization of enhanced green fluorescent protein (eGFP)-labeled microvessels confirmed the human origin of these capillaries. No human vasculature was observed after implantation of HT1080 fibrosarcoma cells, 293 human embryonic kidney cells, or human skin fibroblasts. Intravascular red fluorescent microspheres injected into host circulation were found within green "telomerized" microvessels, indicating functional murine-human vessel anastamoses. Whereas primary HDMEC-derived vessel density decreased with time, telomerized HDMEC maintained durable vessels six weeks after xenografting. Modulation of implant vessel density by exposure to different angiogenic and angiostatic factors demonstrated the utility of this system for the study of human microvascular remodeling in vivo.


Assuntos
Endotélio Vascular/enzimologia , Endotélio Vascular/transplante , Microcirculação , Modelos Animais , Telomerase/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Membrana Basal/química , Capilares/efeitos dos fármacos , Capilares/crescimento & desenvolvimento , Células Cultivadas , Quimera , Colágeno/análise , Colágeno/farmacologia , Colágeno Tipo XVIII , Derme/irrigação sanguínea , Endostatinas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fibroblastos , Humanos , Camundongos , Camundongos SCID , Microcirculação/efeitos dos fármacos , Microcirculação/crescimento & desenvolvimento , Microesferas , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Telomerase/genética , Telômero/genética , Transdução Genética , Transplante Heterólogo , Células Tumorais Cultivadas
4.
J Natl Cancer Inst ; 86(17): 1303-14, 1994 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-7520508

RESUMO

BACKGROUND: Cancer incidence increases with age, but the growth and spread of tumors is often slow and prolonged in the elderly. Transplantable murine tumors grow and spread less readily in older mice and can be used as models to study the effect of host age. Neovascularization is crucial for the growth of solid tumors, and alterations in the host vascular response may underlie the changes in tumor growth occurring with age. PURPOSE: We have used transplantable tumor cells and tumors, and tumor extracts, to better understand differences in the biology of tumor growth and vascularization as a function of host age. METHODS: Englebreth-Holm-Swarm (EHS) carcinoma and B16-F10 melanoma cells were injected into C57BL mice of different ages. Tumor growth, histology, and cellular DNA synthesis were compared. Vascularization was determined using basic fibroblast growth factor and an in vivo angiogenesis assay. EHS tumor extracts were assayed for biologic activity in vitro using human endothelial cells and in vivo using mouse EHS-BAM carcinoma and human TSU-Pr1 prostate carcinoma cells. RESULTS: EHS tumors formed larger tumors in young than in old C57BL mice. Rapid tumor growth resumed upon transfer of tumor tissue from old animals into young animals. The rate of DNA synthesis of tumor tissue from old animals in organ culture was lower than in tissue from young animals. Histologically, tumors grown in old animals exhibited a threefold higher ratio of extracellular matrix to tumor cells than those grown in young animals. Tumors from adult animals exhibited numerous small blood vessels; those from old animals contained fewer, much larger vessels. Similar results were observed in young mice fed a reduced-calorie diet. Young animals elicited a greater and more rapid angiogenic response than old animals. Extracts of tumors grown in old animals failed to support endothelial cell differentiation in culture. Tumor cells injected together with such old extracts showed reduced tumor growth in nude mice. CONCLUSIONS: The rate of growth and morphology of the EHS tumor were altered with age, partly due to a reduced capacity to vascularize the tumors because of a lack of angiogenic factors or the presence of host inhibitors. IMPLICATIONS: Alterations in host factors detected in this tumor model may underlie a variety of age-dependent changes that could influence tumor growth and the repair and regeneration of normal tissue. Reducing the vascularization of tumors represents a potential target to reduce their growth and progression.


Assuntos
Envelhecimento/fisiologia , Carcinoma/fisiopatologia , Melanoma Experimental/fisiopatologia , Neovascularização Patológica/fisiopatologia , Animais , Ingestão de Energia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas
5.
J Natl Cancer Inst ; 84(20): 1582-7, 1992 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-1328660

RESUMO

BACKGROUND: Small-cell lung cancer (SCLC) is a common malignancy that is usually fatal, since it metastasizes and recurs even after aggressive chemotherapy. While the cellular origin of this cancer is not well established, the cells of certain tumors exhibit neuroendocrine markers, including L-dopa decarboxylase. PURPOSE: We designed in vitro and in vivo studies to investigate whether the neuroendocrine features in classic SCLC cell lines were sufficient to make them sensitive to 1-methyl-4-phenylpyridinium (MPP+), a known neurotoxin that destroys nigrostriatal dopaminergic neurons. METHODS: Both classic SCLC cell lines (NCI-H345, NCI-H510, NCI-H187, and NCI-H146) and variant SCLC cell lines (NCI-H417, NCI-H82, NCI-H446, and NCI-H524) were exposed to MPP+ (0-512 microM) for 3 days. Inhibition of DNA synthesis was determined by [3H]thymidine incorporation assays. In a related experiment, MPP+ was removed from the classic cell line culture, and the incorporation of [3H]thymidine was determined. In the in vivo study, male athymic nude mice received subcutaneous injections of 0.5 mL tumor cells with matrigel for 10 days to enhance tumor growth, followed by MPP+ at doses of 100-400 micrograms/d given intraperitoneally for 2 days. RESULTS: All four classic SCLC cell lines showed great sensitivity to MPP+, with detachment from laminin substrates and inhibition of DNA synthesis. MPP+ interfered with [3H]thymidine incorporation and, thus, with DNA synthesis in classic SCLC cell lines at low doses (median +/- SD, 12 +/- 4 microM), whereas much higher doses (median, > 512 microM) were required to inhibit [3H]thymidine incorporation in the variant lines. Treated cells excluded trypan blue dye, showing that inhibition of DNA synthesis was not due to cytotoxicity, and the cells incorporated [3H]thymidine when MPP+ was removed from the culture medium, demonstrating that the inhibition was reversible. MPP+ inhibited the growth of the classic NCI-H187 and variant NCI-H417 cell lines implanted in nude mice. CONCLUSIONS: These results suggest that MPP+ differentially interferes with DNA synthesis in SCLC cell lines in vitro; the selective inhibitory effect on classic cell lines suggests that the neuroendocrine properties expressed by classic SCLC cells may be responsible for the differential effect. IMPLICATIONS: MPP+ exerts a cytostatic effect on these cell lines, and the differential sensitivity observed in vitro is maintained in vivo, suggesting that MPP+ or other pyridinium compounds may be of therapeutic value in SCLC.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Animais , Carcinoma de Células Pequenas/metabolismo , Carcinoma de Células Pequenas/patologia , DNA de Neoplasias/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Timidina/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
6.
Cancer Res ; 61(13): 4994-5001, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431332

RESUMO

Understanding the regulation of endothelial cell (EC) gene expression has important implications for angiogenesis, tumor growth, and metastasis. The transcription factor runt-related gene 2 (RUNX2)/core binding factoralpha-1/acute myeloid leukemia 3/polyoma enhancer-binding protein 2alphaA/osteoblast-specific transcription factor 2 regulates osteoblast differentiation, increases lymphomagenesis in transgenic mice, and is expressed in murine ECs. Here, we report on RUNX2 expression in human bone marrow EC (HBME-1) and its role in EC differentiation. Expression of RUNX2 occurred in HBME-1 cultured on extracellular matrix (ECM) substrates that stimulate in vitro differentiation (tube formation). Neutralizing anti-insulin-like growth factor (IGF)-I-receptor antibody inhibited tube formation as well as activation of RUNX2 expression in HBME-1 cultured on ECM. IGF-I treatment also increased both RUNX2 mRNA and protein expression. HBME-1 transfectants expressing dominant-negative (DN) RUNX were established to address the role of RUNX2 in these processes. HBME/DN cells exhibited reduced tube formation activity relative to control transfectants and less ability to growth arrest and differentiate on ECM. DNRUNX expression also inhibited HBME-1 migration and invasion, which are necessary for tube formation. The urokinase-type plasminogen activator and membrane-type MMP-1 genes were consistently down-regulated in DNRUNX transfectants. The results suggest that RUNX2 is important in IGF-I and ECM-regulated EC migration and differentiation. RUNX2 effects on HBME-1 migration and invasion may occur through activation of protease expression, events that regulate angiogenesis, and tumor growth.


Assuntos
Células da Medula Óssea/fisiologia , Movimento Celular/genética , Endotélio Vascular/fisiologia , Proteínas de Neoplasias , Fatores de Transcrição/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Bovinos , Diferenciação Celular/genética , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core , Subunidades alfa de Fatores de Ligação ao Core , DNA Complementar/genética , Endopeptidases/biossíntese , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Neovascularização Fisiológica/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Ativação Transcricional/genética
7.
Cancer Res ; 50(22): 7261-71, 1990 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-2121334

RESUMO

Surface galactosyltransferase (GT) has been described on a variety of cells where it is believed to be involved in cell-cell and cell-substratum adhesion. Here we show that B16 metastatic murine melanoma cells exhibit a 5-fold higher cell surface GT activity than their nonmetastatic counterparts, although total GT activity in NP-40 solubilized cells is similar for both melanoma variants. Interestingly, on living cells, this cell surface GT almost exclusively galactosylates an endogenous glycoprotein (Mr = 110,000). This metastasis-associated GT is specific for terminal D-N-acetylglucosamine, catalyzes the formation of a beta 1-4 linkage, does not recognize polylactosaminoglycans, and has its specificity altered from D-N-acetylglucosamine to D-glucose by alpha-lactalbumin; yet the Mr = 110,000 protein is not a major substrate when exogenous bovine GT is used on the outside of living cells. In addition to this protein-specific endogenous GT activity, another cell surface GT activity that selectively galactosylates glucosylceramide is also prominent. Endogenous galactosylation of both protein and glycolipid substrates is reduced when the membrane is solubilized by the detergent NP-40 but remains unaltered in the presence of digitonin, which permeabilizes but does not dissolve the membrane. These data suggest that the GTs and their substrates are associated on the cell surface. Chloroquine treatment of intact cells leads to a 4-fold and a 3-fold increase in galactosylation of the Mr = 110,000 protein and glucosylceramide, respectively, suggesting that these two substrates normally reside mostly in the lysosomal or Golgi compartments. The increased expression of lysosomal membrane proteins on the surfaces of highly metastatic cells may, in part, also explain the galactosylation differences observed. These studies further suggest that increased surface localization of certain glycosyltransferases with highly restricted in situ substrate specificities may be a common feature of highly metastatic tumor cells.


Assuntos
Antígenos CD , Gangliosídeo G(M1) , Galactosiltransferases/metabolismo , Lactosilceramidas , Melanoma/enzimologia , Animais , Membrana Celular/metabolismo , Cloroquina/farmacologia , Cromatografia em Camada Fina , Galactose/metabolismo , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Glicoesfingolipídeos/metabolismo , Glicosilação , Isomerismo , Lactalbumina/farmacologia , Melanoma/patologia , Camundongos , Peso Molecular , Metástase Neoplásica , Processamento de Proteína Pós-Traducional , Células Tumorais Cultivadas
8.
Cancer Res ; 55(19): 4210-3, 1995 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-7671222

RESUMO

The use of replication-deficient adenoviral vectors in gene therapy may become a powerful method to achieve efficient but safe transfer of anti-tumor agents. Introduction of the wild-type p53 gene into tumor cells has, in general, been associated with growth suppression. In this study, infection of androgen-independent human prostate Tsu-pr1 cells lacking functional p53 alleles resulted in high levels of p53 protein within 10-15 h. Cells infected with AdCMV.p53 detached from the substratum, condensed, and exhibited fragmentation of nuclear DNA into nucleosomal units consistent with the process of apoptosis. These effects were evident within 24 h after infection, and the majority of cells had undergone apoptosis by 48 h, whereas cells infected with AdCMV.NLS beta Gal continued to proliferate. Uninfected or AdCMV.NLS beta Gal-infected Tsu-pr1 cells formed tumors in nude mice within 3 weeks after implantation, whereas AdCMV.p53-infected cells failed to form tumors during this period. Therefore, adenoviral-mediated antitumor therapy using the p53 gene is an efficient method to inhibit prostate tumor growth, and agents that target the cellular programmed cell death pathway may be useful in clinical applications.


Assuntos
Adenoviridae/genética , Apoptose , Genes p53 , Terapia Genética , Neoplasias da Próstata/prevenção & controle , Animais , Humanos , Masculino , Camundongos , Camundongos Nus , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/análise
9.
Cancer Res ; 57(23): 5328-35, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9393757

RESUMO

Adaptation to hypoxia represents an important aspect of tumor progression. Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that mediates essential homeostatic responses to cellular and systemic hypoxia by activating transcription of multiple genes including those encoding glycolytic enzymes and vascular endothelial growth factor (VEGF). In this report, we demonstrate that whereas C-SRC expression is not required for expression of HIF-1 or transcriptional activation of genes encoding VEGF and enolase 1 (ENO1), cells expressing the v-Src oncogene have increased expression of HIF-1, VEGF, and ENO1 under both hypoxic and nonhypoxic conditions. Expression of V-SRC was associated with increased transcription of reporter genes containing cis-acting hypoxia-response elements from the VEGF and ENO1 genes, and this transcriptional activation required an intact HIF-1 binding site. When three rat hepatoma subclones that differed with respect to the level of HIF-1 expression were injected into nude mice, tumor growth correlated with HIF-1 expression, suggesting that HIF-1 may be generally involved in tumor progression. These studies link an oncogene to the induction of HIF-1 expression, thus providing a mechanism for hypoxic adaptation by tumor cells.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Fatores de Crescimento Endotelial/biossíntese , Neoplasias Hepáticas Experimentais/patologia , Linfocinas/biossíntese , Proteínas Nucleares/biossíntese , Proteína Oncogênica pp60(v-src)/biossíntese , Fosfopiruvato Hidratase/biossíntese , Transcrição Gênica , Células 3T3 , Animais , Sequência de Bases , Linhagem Celular , Células Clonais , Progressão da Doença , Embrião de Mamíferos , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias Hepáticas Experimentais/fisiopatologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Proteínas Proto-Oncogênicas pp60(c-src)/biossíntese , RNA Mensageiro/biossíntese , Ratos , Proteínas Recombinantes/biossíntese , Fatores de Transcrição/biossíntese , Transfecção , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
10.
Cancer Res ; 52(8): 2222-7, 1992 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-1559226

RESUMO

Many previous studies have implicated cell surface saccharides, and sialylglycoconjugates in particular, as important mediators of tumor cell metastasis. In this report, we have used three different specific sialidases and a highly sensitive high-performance liquid chromatographic sialic acid assay to probe the cell surfaces of several murine adrenal carcinoma variants. In contrast to several earlier studies on other metastatic variants, we find no significant differences in the overall levels of cell surface or total cellular sialic acid among three Y1 murine adrenal carcinoma variants with widely different metastatic phenotypes. However, using highly purified, linkage-specific sialyltransferases, in conjunction with V. cholerae sialidase, to probe the cell surface saccharide topography of specific penultimate oligosaccharides, we do find striking differences in oligosaccharide structures underlying the sialic acid moieties. Two tumorigenic and metastatic variants (F2 and F4) contain about 6-fold more penultimate Gal beta 1----4GlcNAc sialylation sites than a related tumorigenic but nonmetastatic variant (HSR) when CMP-[3H]-N-acetylneuraminic acid and the Gal beta 1----4GlcNAc alpha 2,6 sialyltransferase are used to probe the adrenal carcinoma cell surfaces. The metastatic variants also are found to contain 4- to 4.5-fold more Gal beta 1----3GalNAc sialylation sites than the nonmetastatic variant when the Gal beta 1----3GalNAc alpha 2,3 sialyltransferase is used as a cell surface probe. Earlier work, which used the same sialyltransferase probes on sialidase-treated murine melanoma variants (A. Passaniti and G. W. Hart, J. Biol. Chem., 263: 7591-7603, 1988), also showed similar quantitative differences in penultimate structures between metastatic variants. However, in contrast to the adrenal carcinoma cells, the highly metastatic melanoma cells have severalfold lower levels of sialylatable penultimate Gal beta 1----4GlcNAc and Gal beta 1----3GalNAc saccharides compared to their nonmetastatic counterparts. Thus, while the precise structural alterations or surface accessibilities of penultimate saccharides appear to be cell type dependent, these results suggest that pronounced changes in penultimate cell surface sialo-oligosaccharide moieties occur during progression to a malignant phenotype in two widely different tumor systems. These types of alterations in the underlying penultimate oligosaccharide structures of cell surface sialoglycoconjugates may be a common feature of highly metastatic cells arising from very different tumor cell types.


Assuntos
Neoplasias das Glândulas Suprarrenais/química , Glicoproteínas/análise , Proteínas de Neoplasias/análise , Oligossacarídeos/análise , Ácidos Siálicos/análise , Acetilgalactosamina/análise , Acetilglucosamina/análise , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Cromatografia Líquida de Alta Pressão , Metástase Neoplásica , Neuraminidase , Conformação Proteica , Células Tumorais Cultivadas
11.
Cancer Res ; 55(13): 2920-6, 1995 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-7540952

RESUMO

The development of drugs that target the tumor neovasculature may hold promise in inhibiting tumor growth. Experiments in vivo with castanospermine, an inhibitor of the glucosidases that convert protein N-linked high mannose carbohydrates to complex oligosaccharides, resulted in significant inhibition of tumor growth in nude mice. Angiogenesis to basic fibroblast growth factor in castanospermine-treated C57/BL mice was similarly reduced. Endothelial cell proliferation, invasion of basement membrane, and differentiation are crucial steps during neovascularization. In vitro differentiation models using Matrigel and postconfluent cultures of endothelial cells were used to study the effects of glycosidase inhibitors on endothelial cell behavior. FACS analysis of cell surface oligosaccharides using either Concanavalin A or L-phytohemagglutinin lectins confirmed an increase in high mannose groups and a decrease in tri- and tetra antennary beta-linked galactose-N-acetylglucosamine on mannose residues of Asn-linked oligosaccharides upon drug treatment. Castanospermine and the glucosidase inhibitor N-methyldeoxynojirimycin prevented the morphological differentiation of endothelial cells in vitro. These compounds did not alter the proliferation of cultured endothelial cells or their ability to attach to various extracellular matrix molecules. However, the cells showed a reduced ability to migrate and to invade basement membrane gels in vitro and an increased tendency to form aggregates that was inhibitable by D-mannose. These studies suggest that certain cell surface oligosaccharides are required for angiogenesis and that glucosidase inhibitors that alter these structures on endothelial cells are able to inhibit tumor growth.


Assuntos
Endotélio/metabolismo , Glicoconjugados/metabolismo , Inibidores de Glicosídeo Hidrolases , Glicosilação/efeitos dos fármacos , Indolizinas/farmacologia , Neoplasias Experimentais/patologia , Neovascularização Patológica , 1-Desoxinojirimicina/análogos & derivados , Animais , Adesão Celular/efeitos dos fármacos , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Glucosamina/análogos & derivados , Glucosamina/farmacologia , Inibidores do Crescimento , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Processamento de Proteína Pós-Traducional/efeitos dos fármacos
12.
Cancer Res ; 55(2): 399-407, 1995 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-7529136

RESUMO

A hormone-dependent, clonal carcinoma cell line, designated RM22-F5, was derived from a malignant mammary mixed tumor spontaneously arising in an outbred old female Wistar rat. These cells expressed keratin and desmosomal protein and formed epithelial monolayers in a growth factor and hormone-supplemented medium (LHC-8) containing 10% fetal bovine serum (E-type cells). Cells subcultured for 6 to 8 passages in RPMI 1640 medium containing 10% fetal bovine serum without supplements appeared to be fibroblastic and expressed vimentin (F-type cells). The shift to a fibroblast-like morphology was associated with a more malignant phenotype which included rapid, hormone-independent growth and invasive sarcoma-like character in nude mice. F-type cells were no longer able to express their original epithelial phenotype in LHC-8 medium. Cytogenetic analysis revealed that both E- and F-type cells had essentially the same karyotype. Analysis of PCR-amplified DNA further demonstrated a point mutation of the H-ras-1 gene at codon 12 and loss of the normal H-ras-1 allele in both cell types. Genetic tagging of E-type cells with the neomycin-resistance gene resulted in the generation of F-type cells with neomycin resistance in RPMI 1640 medium, suggesting that F-type cells are a malignant variant of E-type cells arising during in vitro culture. Somatic cell fusion between E- and F-type cells revealed that with most hybrid clones tested, the fibroblast-like phenotype was greatly suppressed. These results suggest that an irreversible phenotypic transition, representative of tumor progression from hormone-dependent adenocarcinoma to more malignant hormone-independent spindle carcinoma cells, is a recessive event and may involve loss of a suppressor function.


Assuntos
Adenocarcinoma/patologia , Carcinoma/patologia , Transformação Celular Neoplásica/patologia , Neoplasias Mamárias Animais/patologia , Neoplasias Hormônio-Dependentes/patologia , Adenocarcinoma/química , Adenocarcinoma/genética , Animais , Sequência de Bases , Carcinoma/química , Carcinoma/genética , Divisão Celular , Fusão Celular , Transformação Celular Neoplásica/química , Transformação Celular Neoplásica/genética , Meios de Cultura , Desmossomos/química , Feminino , Citometria de Fluxo , Genes ras/genética , Queratinas/análise , Neoplasias Mamárias Animais/química , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Neomicina , Invasividade Neoplásica , Neoplasias Hormônio-Dependentes/química , Neoplasias Hormônio-Dependentes/genética , Fenótipo , Ratos , Ratos Wistar , Transfecção , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/patologia , Vimentina/análise
13.
Cancer Res ; 53(8): 1833-7, 1993 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-7682157

RESUMO

Linomide (N-phenylmethyl-1,2-dihydro-4-hydroxyl-1-methyl-2-oxoquinoline-3-carboxa mide) has a reproducible in vivo antitumor effect against a series of both androgen responsive and independent Dunning R-3327 rat prostatic cancers. This antitumor effect of linomide is host mediated. One possible mechanism involving the host is that linomide has antiangiogenic activity. An indication that linomide treatment has antiangiogenic activity is the observation that prostatic cancers from linomide treated rats have more focal necrosis than sized matched tumors from untreated rats. To directly test if linomide has antiangiogenic activity, a newly developed Matrigel based quantitative in vivo angiogenic assay was used. These experiments demonstrated that linomide has dose dependent, antiangiogenic activity in vivo in the rat. Additional studies demonstrated that due to its antiangiogenic activity, linomide treatment of rats bearing prostate cancers resulted in a more than 40% decrease in tumor blood flow. Blood flow to a variety of non-tumor bearing organs was not decreased suggesting that linomide selectively inhibits angiogenesis and does not induce loss of established blood vessels. Using as a model the response of human umbilical vein endothelial cells to linomide treatment in a variety of in vitro assays, linomide was demonstrated to have cytostatic but not cytotoxic effect on human umbilical vein endothelial cells at a medium concentration of > or = 100 micrograms/ml. In addition, both endothelial cell chemotactic migration and invasion are steps in angiogenesis inhibited by linomide treatment.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antineoplásicos/farmacologia , Hidroxiquinolinas/farmacologia , Neovascularização Patológica/prevenção & controle , Animais , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Humanos , Masculino , Neoplasias da Próstata/irrigação sanguínea , Ratos , Ratos Sprague-Dawley
14.
Oncogene ; 22(15): 2334-42, 2003 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-12700668

RESUMO

The Ewing's sarcoma family of tumors (ESFT) contain a translocation, t(11;22), which results in the novel oncogenic fusion protein EWS/FLI1. Platelet-derived growth factors (PDGF) and their receptors (PDGFR) are involved in the induction and proliferation of numerous solid tumors and are the potential candidates for novel targeted antitumor therapy. Since a relation was reported between PDGF-C and EWS/FLI1, we sought to characterize the PDGF signaling pathway in ESFT. Eight out of nine ESFT cell lines were found to express significant levels of beta-PDGFR. Interestingly, none of the tested cell lines expressed alpha-PDGFR, which is the receptor isotype required for PDGF-C binding. By immunohistochemical staining 47 of 52 (90.4%) archival tumor samples from patients with ESFT were positive for beta-PDGFR. ESFT cell lines were treated with PDGF-AA or PDGF-BB ligands to evaluate downstream signaling. Autophosphorylation of beta-PDGFR and tyrosine phosphorylation of PLC-gamma, PI3Kp85 and Shc were detected only in PDGF-BB-stimulated cells that express beta-PDGFR. Receptor function was further evaluated using chemotaxis assays that showed TC-32 cell migration towards PDGF-BB. A specific PDGFR kinase inhibitor AG1295 blocked beta-PDGFR activation, downstream signaling, growth in cell culture and chemotaxis of TC-32 cells. AG1295 also delayed tumor formation and prolonged survival in an ESFT animal model. We conclude that ESFT express beta-PDGFR and that this is a functional and potentially crucial signaling pathway. Therefore, beta-PDGFRs may provide a novel therapeutic target in ESFT that can be utilized to design better treatment modalities.


Assuntos
Neoplasias Ósseas/patologia , Proteínas de Neoplasias/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Sarcoma de Ewing/patologia , Animais , Becaplermina , Neoplasias Ósseas/metabolismo , Divisão Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Classe Ib de Fosfatidilinositol 3-Quinase , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Humanos , Isoenzimas/metabolismo , Camundongos , Camundongos SCID , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor beta de Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Recombinantes/farmacologia , Sarcoma de Ewing/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/metabolismo , Fosfolipases Tipo C/metabolismo , Tirfostinas/farmacologia
15.
Hum Gene Ther ; 6(11): 1457-65, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8573618

RESUMO

In vivo gene transfer of angiogenic growth factors represents a potential approach to the treatment of ischemic diseases. The present study examined the in vitro and in vivo effects of two replication-deficient recombinant adenovirus (Ad) vectors coding for human acidic fibroblast growth factor (aFGF1-154). One vector codes for the nonsecreted form of the peptide (AdCMV.aFGF1-154), and the other vector codes for a recombinant, secreted form (AdCMV.sp+aFGF1-154). AdCMV.NLS beta gal, an adenovirus vector coding for beta-galactosidase (beta-Gal), was used as a control. Assessment of proliferation of starved human umbilical vein endothelial cells infected with AdCMV.aFGF1-154 and AdCMV.sp+aFGF1-154 (20 pfu/cell) showed approximately 6- and 10-fold increase in cell number over control, respectively. Infection with AdCMV.sp+aFGF1-154 and with AdCMV.aFGF1-154 enhanced endothelial cell differentiation into capillary-like structures in vitro. However, this effect was significantly more pronounced with AdCMV.sp+aFGF1-154 than with AdCMV.aFGF1-154. Angiogenesis in vivo was assessed by injecting subcutaneously into mice 750 microliters of reconstituted basement membrane proteins (Matrigel) and the Ad vectors (2 x 10(8) pfu). After 14 days, there was histologic evidence of neovascularization in the animal's tissue surrounding the Matrigel plugs with AdCMV.aFGF1-154 and AdCMV.sp+aFGF1-154. Further, the hemoglobin content of the Matrigel plugs with AdCMV.aFGF1-154 and with AdCMV.sp+aFGF1-154 was, respectively, 2.3- and 2.6-fold higher than with AdCMV.NLS beta gal. Together, these observations support the concept that adenovirus vectors coding for various forms of acidic FGF1-154 may be used to induce angiogenesis in vivo and may provide a new therapeutic approach to ischemic diseases.


Assuntos
Adenoviridae/genética , Fator 1 de Crescimento de Fibroblastos/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Diferenciação Celular , Divisão Celular , Linhagem Celular , Células Cultivadas , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL
16.
Matrix Biol ; 14(7): 561-71, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8535606

RESUMO

During development of the embryonic limb, differentiation of mesenchymal progenitor cells into chondrocytes is regulated by cell shape, extracellular matrix, and growth and differentiation factors. In this study, reconstituted basement membrane (Matrigel) prepared from mouse Englebreth-Holm-Swarm tumor tissue was found to stimulate mesenchymal cell chondrogenesis in vitro and the production of cartilage at ectopic sites in athymic mice. The rate of chondrogenesis of mesenchymal cells from chick limb bud was increased four-fold by the addition of 400 micrograms/ml Matrigel to the media of micromass cultures, and this activity was not blocked by neutralizing antibodies to transforming growth factor-beta (TGF-beta) or fibroblast growth factor (FGF). Mesenchymal cells cultured on Matrigel, but not laminin or collagen type I or IV, formed spheres of condensed cells which stained with Alcian blue. Chick limb-bud mesenchymal cells suspended in Matrigel prepared from tumors grown in C57 mice aged 3, 12, or 26 months formed disks of hyaline cartilage within 2 weeks with wet weights of 59.1 mg, 35.7 mg, and 21.4 mg, indicating that the Matrigel from the old animals was less biologically active. In agreement with the in vivo data, Alcian blue staining of proteoglycan was over two-fold higher in micromass cultures supplemented with the Matrigel from young animals than in cultures treated with the Matrigel from old mice. A high-salt wash preparation of Matrigel from tumors grown in old mice increased the rate of chondrogenesis and cartilage production, suggesting that an inhibitor of chondrogenesis is produced by the old host. Thus, Matrigel contains chondrogenic activity distinct from TGF-beta or FGF. The aged host may produce factors that are inhibitory to mesenchymal cell differentiation and adversely affect cartilage formation and repair.


Assuntos
Membrana Basal/fisiologia , Cartilagem/embriologia , Desenvolvimento Embrionário e Fetal , Matriz Extracelular/fisiologia , Extremidades/embriologia , Mesoderma/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Embrião de Galinha , Colágeno/farmacologia , Combinação de Medicamentos , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Laminina/farmacologia , Mesoderma/citologia , Camundongos , Camundongos Nus , Proteoglicanas/farmacologia , Sarcoma Experimental/metabolismo , Fatores de Tempo
17.
Mech Ageing Dev ; 106(1-2): 93-102, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9883975

RESUMO

Loss of methylation at a CpG island in exon I of the rat ER gene was observed in 48% of the spontaneous mammary tumors in old female Wistar rats and 22% of the contralateral normal mammary tissues. The majority of the methylation losses were total. Similarly, 50% of 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary tumors in young Sprague-Dawley rats exhibited a partial or total loss of methylation at this site, while all normal mammary tissues in young rats were fully methylated. Loss of ER methylation also increased with age in normal mammary tissues of tumor-free rats approaching 12.5% in middle-aged and 43% in old rats. In addition, 66% of mammary glands obtained from young rats that are subsequently at an increased risk to develop breast cancer due to manipulation of in utero dietary fat intake, exhibited methylation loss while no methylation changes were observed in rats at no increased risk for breast cancer. Therefore, the loss of ER methylation is more extensive in mammary glands of rats at high than low breast cancer risk, in old than young, and in mammary tumors than in normal tissues. The data suggest that hypomethylation of a growth-associated ER gene may be a common event in mammary tumorigenesis in the rat and may be of predictive value as a marker of increased breast cancer risk in aged individuals.


Assuntos
Envelhecimento/genética , Ilhas de CpG , Metilação de DNA , Éxons , Neoplasias Mamárias Experimentais/genética , Receptores de Estrogênio/genética , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Testes de Carcinogenicidade , Carcinógenos/farmacologia , Linhagem Celular , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Ratos Wistar
18.
Biotechniques ; 24(6): 1038-43, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9631200

RESUMO

Screening methods for chemotherapeutic agents usually rely on the cytotoxic properties of the drugs. However, agents that inhibit invasion may have more efficacy and cause fewer side effects. Various cellular invasion assays have been used to evaluate these types of compounds, including the modified Boyden chamber, monolayer wound models and Matrigel outgrowth assays. In this report, we have combined the use of the Matrigel outgrowth assay with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) visualization and cell viability dye to visualize invasive cells on Matrigel without magnification. Extraction of the dye's formazan byproduct allows cell viability to be assessed. Using several invasive and noninvasive cell lines, the utility of the method for various target cells was verified. Several established chemotherapeutic agents were also screened for their anti-invasive and/or cytotoxic effects when cultured on Matrigel. Our results suggest that this method may be an easy, inexpensive and nonradioactive alternative for both enumerating cells on Matrigel and screening various tumor cell lines treated with chemotherapeutic agent to look for compounds with noncytotoxic but anti-invasive properties.


Assuntos
Antineoplásicos/química , Colágeno , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Laminina , Invasividade Neoplásica/patologia , Proteoglicanas , Células Tumorais Cultivadas/patologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Combinação de Medicamentos , Feminino , Humanos , Masculino , Sais de Tetrazólio , Tiazóis
19.
Cancer Lett ; 100(1-2): 47-54, 1996 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-8620453

RESUMO

Primary prostate epithelial and prostate adenocarcinoma cells cultured in serum-free medium grew for up to 10 passages before senescence. Cells from prostate adenocarcinoma of a 55-year-old patient without lymph node involvement were transfected with plasmids containing recombinant human papilloma virus HPV16 or HPV18 DNA and the selectable neomycin-resistance gene. After G-418 selection, cells underwent crisis, and surviving cells infected with retroviruses encoding the HPV18 E6/E7 genes (HPV-PAC1), transfected with a head-to-tail dimer of the complete HPV16 genome (HPV-PAC2), or transfected with HPV18 E6/E7 early genes (HPV-PAC3) were established. HPV-PAC1 and HPV-PAC2 cultures appeared morphologically similar to primary cultures even after 40 passages. However, HPV-PAC2 cultures had a clonal morphology. All lines were positive for cytokeratin 18, had acquired vimentin expression, and contained either HPV16 or HPV18 sequences integrated into host DNA. None was tumorigenic in nude mice or formed colonies in soft agar. These cells did not secrete prostate specific antigen nor respond to androgen although tamoxifen inhibited the growth of the cells. Immunohistochemistry showed no evidence of p53 overexpression. Further characterization of these cell lines and examination of their response to chemotherapeutic agents may provide relevant information for the study of hormone-independent PC.


Assuntos
Adenocarcinoma/virologia , DNA Viral/genética , Papillomaviridae/genética , Neoplasias da Próstata/virologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Androgênios/farmacologia , Divisão Celular/efeitos dos fármacos , Meios de Cultura , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Estimulação Química , Transfecção , Células Tumorais Cultivadas
20.
Exp Gerontol ; 27(5-6): 559-66, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1426088

RESUMO

The incidence of many cancers shows a sharp increase with age. This is particularly true of prostate cancer, which arises in many older males. Little or no morbidity is observed as the tumor develops in situ in the prostate. However, once malignant cells escape from the primary lesion and metastasize, the disease assumes a much more serious course. Here we report on the activity of human prostate cancer cells in culture as well as their behavior when transplanted into nude mice. In vitro, several lines of prostate carcinoma cells obtained from metastatic lesions were embedded in reconstituted basement membrane proteins (Matrigel) and found to exhibit highly invasive activity as observed with malignant cells from other types of tumors. Also, we report an improved method for obtaining an increased growth of human prostate cancer cells in nude mice by injecting these cells in Matrigel. Since there are no adequate animal models of prostate cancer, the systems described here may prove useful in defining events underlying the development and progression of the tumor cells to malignant status, as well as facilitate the analyses of novel therapeutic agents to prevent the growth and the spread of this cancer.


Assuntos
Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Animais , Divisão Celular , Movimento Celular , Colágeno , Combinação de Medicamentos , Humanos , Laminina , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteoglicanas , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA