Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncogene ; 27(26): 3729-38, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18223682

RESUMEN

Antiangiogenic therapies are promising approaches to cancer control, but the details of their effects on subsequent tumor progression are not fully understood. Such therapies have the potential to eventually generate extensive amounts of tumor ischemia, and we previously demonstrated that ischemic conditions induce K-ras mutations in cells with deficient mismatch repair (MMR) mechanisms. This suggested that similar effects on oncogene mutagenesis may accompany antiangiogenic therapy. To test this, MMR-deficient colorectal cancer cells (Dks-8) were xenografted into immune-deficient mice and treated with the antiangiogenic regimen of low-dose/metronomic cyclophosphamide for 2 weeks followed by a 2-week recovery period without therapy. This treatment resulted in transient tumor growth inhibition, increased hypoxia, and decreased microvessel density, and cancer cells from treated tumors acquired activating mutations of the K-ras oncogene (K-ras(G13D)). In vitro exposure of Dks-8 cells to the active metabolite of cyclophosphamide (4-hydroxycyclophosphamide) had no effect on the K-ras status, indicating that there was no direct action of this alkylating agent on K-ras mutagenesis. In addition, cells sorted from hypoxic regions of Dks-8 tumors were enriched in K-ras(G13D) mutants. Collectively, our studies suggest that increases in tumor hypoxia induced by antiangiogenic treatment may lead to K-ras mutation and consequently tumor progression, especially in susceptible individuals.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Hipoxia de la Célula , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Ciclofosfamida/uso terapéutico , Genes ras , Mutación , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/metabolismo , Ciclofosfamida/análogos & derivados , Humanos , Ratones , Trasplante de Neoplasias , Trasplante Heterólogo
3.
Am J Pathol ; 158(4): 1325-34, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11290550

RESUMEN

Cells within a tumor are highly heterogeneous with respect to a wide range of genotypic and phenotypic characteristics. The latter include such properties as growth, survival, invasion, and metastasis. We asked whether the degree to which individual tumor cells rely on a tumor's vasculature might also be heterogeneous. By adapting an intravital Hoechst 33342 staining technique, we labeled and isolated tumor cells based on their relative proximity to perfused vessels. Because tumor regions distal to the vasculature are likely hypoxic, we examined cells deficient for hypoxia-inducible factor-1alpha (HIF-1alpha), a transcription factor that has been shown to mediate hypoxia-induced responses, including apoptosis. Despite reduced vascularization in HIF-1alpha-/- embryonic stem cell-derived tumors, their growth in vivo was found to be accelerated relative to HIF-1alpha+/+ tumor counterparts. We hypothesized that this paradoxical observation is because of decreased apoptotic rate, resulting in diminished vascular dependence of HIF-1alpha-/- cells. Analysis of heterogeneous tumors established from mixtures of HIF-1alpha+/+ with HIF-1alpha-/- cells revealed that the proportion of cells expressing wild-type HIF-1alpha was increased in perivascular areas and decreased in distal tumor regions. Thus, cells expressing HIF-1alpha were found to be highly dependent on proximity to blood vessels for their growth and survival in vivo, whereas cells that had lost HIF-1alpha expression were much less so. Heterogeneity in angiogenesis dependence was also observed among cell subpopulations isolated from human melanoma xenografts. This potential for selection of less vascular-dependent tumor cell variants throughout the course of disease progression may have important implications for the long-term efficacy of anti-angiogenic therapy.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/fisiopatología , Factores de Transcripción , Animales , Bencimidazoles , Vasos Sanguíneos/fisiopatología , División Celular , Proteínas de Unión al ADN/metabolismo , Colorantes Fluorescentes , Genotipo , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Melanoma/irrigación sanguínea , Melanoma/patología , Melanoma/fisiopatología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias/patología , Proteínas Nucleares/metabolismo , Teratoma/irrigación sanguínea , Teratoma/patología , Teratoma/fisiopatología , Trasplante Heterólogo
4.
J Cell Biol ; 142(2): 557-71, 1998 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-9679152

RESUMEN

Recent studies have demonstrated the importance of E-cadherin, a homophilic cell-cell adhesion molecule, in contact inhibition of growth of normal epithelial cells. Many tumor cells also maintain strong intercellular adhesion, and are growth-inhibited by cell- cell contact, especially when grown in three-dimensional culture. To determine if E-cadherin could mediate contact-dependent growth inhibition of nonadherent EMT/6 mouse mammary carcinoma cells that lack E-cadherin, we transfected these cells with an exogenous E-cadherin expression vector. E-cadherin expression in EMT/6 cells resulted in tighter adhesion of multicellular spheroids and a reduced proliferative fraction in three-dimensional culture. In addition to increased cell-cell adhesion, E-cadherin expression also resulted in dephosphorylation of the retinoblastoma protein, an increase in the level of the cyclin-dependent kinase inhibitor p27(kip1) and a late reduction in cyclin D1 protein. Tightly adherent spheroids also showed increased levels of p27 bound to the cyclin E-cdk2 complex, and a reduction in cyclin E-cdk2 activity. Exposure to E-cadherin-neutralizing antibodies in three-dimensional culture simultaneously prevented adhesion and stimulated proliferation of E-cadherin transfectants as well as a panel of human colon, breast, and lung carcinoma cell lines that express functional E-cadherin. To test the importance of p27 in E-cadherin-dependent growth inhibition, we engineered E-cadherin-positive cells to express inducible p27. By forcing expression of p27 levels similar to those observed in aggregated cells, the stimulatory effect of E-cadherin-neutralizing antibodies on proliferation could be inhibited. This study demonstrates that E-cadherin, classically described as an invasion suppressor, is also a major growth suppressor, and its ability to inhibit proliferation involves upregulation of the cyclin-dependent kinase inhibitor p27.


Asunto(s)
Cadherinas/fisiología , Proteínas de Ciclo Celular , División Celular/fisiología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Supresoras de Tumor , Animales , Cadherinas/genética , Cadherinas/inmunología , Adhesión Celular/fisiología , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/fisiopatología , Ratones , Pruebas de Neutralización , Fosforilación , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/metabolismo , Transfección , Células Tumorales Cultivadas
5.
Proc Natl Acad Sci U S A ; 95(7): 3609-14, 1998 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-9520413

RESUMEN

Targeted disruption of the single mutant K-ras allele in two human colorectal carcinoma cell lines (DLD-1 and HCT-116) leads to loss of tumorigenic competence in nude mice with retention of ability to grow indefinitely in monolayer culture. Because expression of the mutant K-ras oncogene in these cell lines is associated with marked up-regulation of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF), we sought to determine whether this potent angiogenesis inducer plays a role in K-ras-dependent tumorigenic competence. Transfection of a VEGF121 antisense expression vector into DLD-1 and HCT-116 cells resulted in suppression of VEGF/VPF production by a factor of 3- to 4-fold. The VEGF/VPF-deficient sublines, unlike the parental population or vector controls, were profoundly suppressed in their ability to form tumors in nude mice for as long as 6 months after cell injection. In contrast, in vitro growth of these sublines was unaffected, thus demonstrating the critical importance of VEGF/VPF as an angiogenic factor for HCT-116 and DLD-1 cells. Transfection of a full-length VEGF121 cDNA into two nontumorigenic mutant K-ras knockout sublines resulted in a weak but detectable restoration of tumorigenic ability in vivo in a subset of the transfectants, with no consistent change in growth properties in vitro. The findings indicate that mutant ras-oncogene-dependent VEGF/VPF expression is necessary, but not sufficient, for progressive tumor growth in vivo and highlight the relative contribution of oncogenes, such as mutant K-ras, to the process of tumor angiogenesis.


Asunto(s)
Carcinoma/irrigación sanguínea , Carcinoma/genética , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/genética , Factores de Crecimiento Endotelial/genética , Regulación Neoplásica de la Expresión Génica , Genes ras , Linfocinas/genética , Neovascularización Patológica/genética , Animales , Humanos , Ratones , Mutación , Células Tumorales Cultivadas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
6.
Nat Med ; 2(11): 1204-10, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8898746

RESUMEN

A low proliferating fraction in solid tumors limits the effectiveness of cell cycle-dependent chemotherapeutic agents. To understand the molecular basis of such "kinetic" resistance we cultured tumor cells as multicellular spheroids and examined levels of p27Kip1, a cyclin-dependent kinase inhibitor known to be upregulated by intercellular contact in normal cells. When transferred from monolayer to three-dimensional culture, a consistent upregulation (up to 15-fold) of p27 protein was observed in a panel of mouse and human carcinoma cell lines. Antisense-oligonucleotide-mediated downregulation of p27 in EMT-6 mammary tumor cell spheroids reduced intercellular adhesion, increased cell proliferation, sensitized tumor cells to 4-hydroperoxycyclophosphamide, and restored drug- or radiation-induced cell-cycle perturbations repressed in spheroid culture. Our results implicate p27 as a regulator of drug resistance in solid tumors and suggest that tumor-targeted p27 antagonists may be useful chemosensitizers in conjunction with conventional anticancer therapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Proteínas de Ciclo Celular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/metabolismo , Resistencia a Antineoplásicos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Supresoras de Tumor , Animales , Adhesión Celular , División Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Humanos , Ratones , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas
7.
J Natl Cancer Inst ; 88(18): 1285-96, 1996 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-8797768

RESUMEN

BACKGROUND: De novo or acquired resistance to chemotherapeutic drugs continues to be one of the most important obstacles hindering the successful treatment of cancer patients. Consequently, enhancing the efficacy of conventional chemotherapeutic drugs has become an important research goal. Our previous studies using the mouse EMT-6 mammary carcinoma selected for resistance to various alkylating agents in vivo demonstrated that such acquired drug resistance may be manifested in vitro only in cells growing in a three-dimensional configuration but not in conventional monolayer culture. We also found that this phenomenon, which we refer to as "acquired multicellular resistance," is associated with an increase in intercellular adhesion or compaction of the alkylating agent-resistant cell lines grown as aggregates in three-dimensional culture. PURPOSE: The present study further investigates the impact of three-dimensional architecture on acquired multicellular drug resistance and its influence on cell cycle kinetics, cell cycle arrest, and cell survival. METHODS: To test the hypothesis that an increase in three-dimensional compaction is related to the drug resistance properties of the cells, we did the following: 1) selected clones of the EMT-6 cell line that spontaneously formed tightly or loosely adherent aggregates and assessed their respective drug resistance properties in vitro; 2) assayed tumorigenic potential of the tight and loose clones after exposure to defined concentrations of the activated form of cyclophosphamide, 4-hydroperoxycyclophosphamide (4-HC) in vitro; and 3) treated the tight clones with hyaluronidase, an agent capable of disrupting EMT-6 spheroids, and assayed what effect this treatment had on chemosensitivity. We used fluorescence-activated cell sorter analysis to monitor any potential alterations in cell cycle kinetics. RESULTS: The increase in compaction in three-dimensional culture was sufficient to confer resistance to 4-HC. This increase in intercellular adhesion was also associated with a lower proliferating fraction of tumor cells and with an almost completely diminished ability of the cells to arrest in the G2/M phase of the cell cycle after drug exposure. Furthermore, these changes were detectable only in three-dimensional culture, not in conventional monolayer culture. In conventional monolayer culture, all cell types consistently showed a high level of proliferation and arrested in G2/M after exposure to 4-HC. Moreover, hyaluronidase was able to disrupt intercellular adhesion and chemosensitize tumor cells both in vitro and in vivo in an ascites model. CONCLUSION: Earlier studies have demonstrated that hyaluronidase is able to sensitize tumor cells to various anticancer agents. Our studies now demonstrate that this sensitization can occur by a mechanism independent of increased drug penetration. This mechanism is likely to be related to the "anti-adhesive" effect of hyaluronidase, which overrides cell contact-dependent growth inhibition, recruits cells into the cycling pool, and renders tumor cells more sensitive to cytotoxic agents that preferentially kill rapidly dividing cells. IMPLICATIONS: Other tumor-specific "anti-adhesives" should be explored that can be effective chemosensitizers when used in combination with cell cycle-specific drugs for the treatment of small, solid tumors.


Asunto(s)
Antineoplásicos/farmacología , Moléculas de Adhesión Celular/metabolismo , Ciclofosfamida/análogos & derivados , Hialuronoglucosaminidasa/fisiología , Neoplasias Mamarias Experimentales/fisiopatología , Animales , Bromodesoxiuridina/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/fisiología , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclofosfamida/farmacología , Resistencia a Antineoplásicos , Femenino , Citometría de Flujo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo , Células Tumorales Cultivadas
8.
Clin Cancer Res ; 2(8): 1417-25, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9816316

RESUMEN

We have previously shown that a majority of human melanoma cell lines derived from early-stage lesions were growth inhibited by exogenous interleukin 6 (IL-6) in vitro, whereas cell lines from advanced-stage lesions were resistant to such IL-6-induced growth inhibition. Among the resistant melanoma cell lines, 50-60% constitutively produced IL-6, which appeared to function as a growth stimulator in vitro, based on the growth-suppressive effects of antisense oligonucleotides to the IL-6 gene. The present study was primarily aimed at evaluating whether endogenous IL-6 also functions in vivo as a growth modulator for IL-6-producing and -nonproducing melanoma cells. To do so, we first introduced an IL-6 expression vector into IL-6-nonproducing human melanoma cells using WM35, an early-stage (radial growth phase) cell line, the growth of which is normally inhibited by IL-6, and WM983A, an advanced-stage cell line, the growth of which in vitro is not affected by exogenous IL-6. None of the IL-6-producing transfectants showed a significant alteration in tumor growth in nude mice. Next, two IL-6-producing melanoma cell lines, both of which were derived from metastases, MeWo and WM9, and which are growth resistant to exogenously added IL-6, were transfected with an antisense IL-6 expression vector. Several transfectant clones manifested a constitutive decrease in IL-6 gene expression and protein production, and they also gave rise to much smaller tumors with slower growth rates and longer latency periods. However, these IL-6 antisense transfectants were not growth suppressed in in vitro cell cultures, relative to their respective parental controls. Taken together, the results demonstrate that endogenous IL-6 can indeed function as a growth stimulator for human cutaneous melanomas in vivo. This growth-stimulatory or survival mechanism remains to be clarified but may be paracrine rather than autocrine in nature.


Asunto(s)
Sustancias de Crecimiento/fisiología , Interleucina-6/fisiología , Melanoma/patología , Animales , Regulación hacia Abajo , Humanos , Interleucina-6/genética , Ratones , Ratones Desnudos , Neovascularización Patológica , Oligonucleótidos Antisentido/farmacología , Células Tumorales Cultivadas
9.
Anticancer Drugs ; 6(1): 3-18, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7538829

RESUMEN

The growth of solid tumors to a clinically relevant size is dependent upon an adequate blood supply. This is achieved by the process of tumor stroma generation where the formation of new capillaries is a central event. Progressive recruitment of blood vessels to the tumor site and reciprocal support of tumor expansion by the resulting neovasculature are thought to result in a self-perpetuating loop helping to drive the growth of solid tumors. The development of new vasculature also allows an 'evacuation route' for metastatically-competent tumor cells, enabling them to depart from the primary site and colonize initially unaffected organs. Several molecular and cellular mechanisms have been identified by which tumor parenchyma may exert its angiogenic effect on host endothelial cells. As a result of this paracrine influence, tumor-associated endothelial cells acquire an 'immature' phenotype manifested by rapid proliferation, migration, release of proteases and expression of cytokines, endothelial-specific tyrosine kinases (e.g. flk-1, tek and others) as well as numerous other molecular alterations. Consequently a network of structurally and functionally aberrant blood vessels is formed within the tumor mass. There is also evidence that endothelial cells themselves, and likewise other stromal cells, may act reciprocally to alter the behavior of adjacent tumor cells in a paracrine or cell contact mediated fashion. For example, production of interleukin 6(IL-6) by endothelial cells may have a differential effect on human melanoma cells expressing different degrees of aggressiveness. In this manner endothelial derived cytokines could conceivably contribute to tumor progression by suppressing the growth of the less aggressive tumor cells and promoting dominance of their malignant counterparts in 'strategic' perivascular zones. Distinct biological features expressed by tumor-associated vasculature may serve as potential prognostic markers of disease progression as well as novel targets for therapeutic intervention.


Asunto(s)
Antineoplásicos/uso terapéutico , Metástasis de la Neoplasia/fisiopatología , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Animales , Antineoplásicos/farmacología , Progresión de la Enfermedad , Diseño de Fármacos , Endotelio Vascular/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/irrigación sanguínea , Melanoma/fisiopatología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias/fisiopatología , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/fisiopatología , Neovascularización Patológica/tratamiento farmacológico , Pronóstico , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/fisiopatología
10.
J Cell Physiol ; 159(2): 245-55, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-8163565

RESUMEN

Tumor progression is frequently associated with changes in responsiveness of tumor cells to paracrine growth factors. A potential major source of such paracrine factors in solid tumors are endothelial cells since this type of cell can constitute a sizeable fraction of the cellular composition of solid tumors. As an initial step to examining the possible effects of endothelial cell-associated growth factors on tumor cell growth, a panel of human melanoma cell lines representative of different stages of tumor progression was employed for studies utilizing endothelial cell-derived growth modulators. Macrovascular or microvascular human endothelial cells from umbilical vein or from skin, respectively, inhibited melanoma cell growth in direct coculture experiments. The potency of this inhibitory effect diminished as a function of melanoma progression. Conditioned media from endothelial cell cultures mimicked the effect of the cell coculture experiments, suggesting the involvement of soluble growth factor(s). Approximately 50-75% of the conditioned media inhibitory effect was abrogated by addition of the neutralizing antibody to interleukin-6 (IL-6). Gel filtration chromatography revealed the presence of additional inhibitors in endothelial cell conditioned medium. Two peaks of activity were detected with apparent molecular weights of approximately 100-150 Kd and 20-30 Kd, the latter containing IL-6 activity. Whereas early-stage radial growth phase (RGP) primary tumor-derived melanoma cells were sensitive to at least three different endothelial products of high or low molecular weight (including IL-6), melanoma cells from more advanced metastatic lesions were resistant to the latter activities, and retained only partial sensitivity to the high molecular weight inhibitor. More advanced vertical growth phase (VGP) primary melanoma cell lines expressed intermediate inhibition-sensitive phenotypes. Thus human melanoma development appears to be associated with progressive loss of sensitivity to the growth inhibitory effects of IL-6 and other factors produced by endothelial cells. This is likely to be a result of a selection process when tumor cells are confronted with adjacent vasculature during the progress of tumor angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/farmacología , Melanoma/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Cromatografía en Gel , Medios de Cultivo Condicionados/farmacología , Resistencia a Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Humanos , Técnicas Inmunológicas , Melanoma/patología , Melanoma/fisiopatología , Estadificación de Neoplasias
11.
In Vitro Cell Dev Biol Anim ; 29A(9): 742-8, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8407718

RESUMEN

In previous experiments it was shown that injection into syngeneic CBA/J mice of cell mixtures containing an excess of non-metastatic SP1 mouse mammary carcinoma cells with a ras transfected metastatic variant of SP1 called C1, always resulted in the eventual dominance of the C1 subpopulation at the site of inoculation. This occurred despite the growth rates of the two cell populations being identical in vivo when grown separately. The means by which the C1 subpopulation achieved "clonal dominance" is thought to involve its responsiveness to stimulatory paracrine growth factors liberated by the non-metastatic SP1 population. The clonal dominance process, however, could not be recapitulated in conventional monolayer tissue culture conditions in which SP1 and C1 cells were grown together in high concentrations of serum, i.e. under non-limiting culture conditions. We now show that clonal dominance of C1 cells can be observed when the cell mixture is maintained in tissue culture for extended periods, or when the cells are grown under selective, limiting conditions, some of which may mimic growth conditions in vivo more accurately. These conditions were a) growth in low (limiting) serum concentrations; and b) growth as three-dimensional multicellular aggregates, i.e. as "tumor spheroids". Under all of these conditions dominance of the C1 subpopulation always took place, but with an efficiency 6- to 40-fold less than generally observed in vivo. C1 cells were also able to form more stable (compact) spheroids compared to SP1 cells. Entrapment of the latter in mixed C1/SP1 spheroids increased the recovery of the SP1 cells suggesting some kind of "rescue" mechanism in which cells are protected from physical forces by three-dimensional structure. The relevance of these in vitro interactions for clonal dominance in primary tumors and metastasis in vivo are discussed.


Asunto(s)
Técnicas de Cultivo/métodos , Metástasis de la Neoplasia/patología , Células Tumorales Cultivadas/citología , Adenocarcinoma/patología , Animales , División Celular , Transformación Celular Neoplásica/patología , Células Clonales/citología , Cinética , Ratones , Ratones Endogámicos CBA , Modelos Biológicos
12.
Cancer Res ; 53(12): 2708-11, 1993 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8504408

RESUMEN

Human melanomas can become progressively resistant to the growth-inhibitory effects of a broad family of structurally diverse cytokines which includes interleukin 6 (IL-6). Uncovering this multicytokine resistance was made possible by the availability of cell lines established from early-stage radial growth phase or vertical growth phase primary melanomas as well as more advanced primary lesions and distant metastases. Because Oncostatin M (OSM) is also a member of the IL-6 family we evaluated the effects of this cytokine on the growth of human melanoma cell lines obtained from different stages of disease progression. The results showed that three different cell lines derived from early-stage melanomas were strongly growth inhibited by OSM, as they are by IL-6. Three cell lines, established from advanced-stage melanomas, were growth inhibited by OSM, but much higher concentrations (in the range of 10-fold) were required to obtain 50% growth inhibition; these cell lines were not inhibited by IL-6. Three other cell lines that were IL-6 resistant (two of which were advanced stage) were also found to be OSM resistant. Only one advanced-stage IL-6-resistant cell line was found to be highly sensitive to OSM-mediated growth inhibition. In addition, we found that variants isolated from early-stage WM35 melanoma cells that possess a much more aggressive tumorigenic phenotype in nude mice were significantly more resistant to both OSM- and IL-6-mediated growth inhibition. The results demonstrate that OSM can function as a growth inhibitor of human melanoma cells but that its ability to do so is progressively diminished or lost with disease progression. This finding is consistent with the concept of acquired "multicytokine resistance" during melanoma progression.


Asunto(s)
Melanoma/patología , Péptidos/farmacología , División Celular/efectos de los fármacos , Resistencia a Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Inhibidores de Crecimiento/farmacología , Humanos , Interleucina-6/farmacología , Factor Inhibidor de Leucemia , Linfocinas/farmacología , Estadificación de Neoplasias , Oncostatina M , Células Tumorales Cultivadas
13.
Br J Cancer ; 65(5): 641-8, 1992 May.
Artículo en Inglés | MEDLINE | ID: mdl-1586590

RESUMEN

A sequential, quantitative loss of Peanut agglutinin (PNA) binding with progression of mouse mammary cells from normal to preneoplastic to neoplastic phenotypes was observed. Normal mammary epithelium, preneoplastic mammary lesions designated D2HAN (D2-type hyperplastic alveolar nodules) and a series of nine spontaneous tumours (D2ST1, D2ST2, D2ST3, D2ST4, D2A1, D2F2, D2.0R, D2.1, EMT6R08) derived from mice bearing D2HAN were grown in culture and analysed by flow cytometry with respect to PNA binding intensity to the cell surface. Primary cultures of normal mammary epithelium strongly bound PNA. A stepwise decrease in PNA binding by preneoplastic D2HAN cells and subsequent tumours arising from those hyperplastic lesions was observed. Three cloned tumour subpopulations derived from such tumours exhibited dramatic differences in PNA binding ranging from high (D2.0R) to low (D2.1) to very low (D2A1 cells). Their growth rate in vitro was similar. However, an inverse correlation between PNA binding and malignant characteristics, such as the incidence and latency of subcutaneous tumours and the efficiency of the tumour cells to form lung colonies after i.v. injection, existed. Cells subsequently derived from tumours resulting from injection of the D2.0R clone (high PNA binding, low tumorigenicity) were found to have diminished PNA binding properties and to be more tumorigenic when reimplanted into syngeneic mice. The difference in PNA binding (up to 50-fold) between normal mammary cells and other mouse mammary tumour cells, i.e., unrelated to D2HAN lesions, was also seen. These include six sister subpopulations derived from a single BALB/cfC3H mouse mammary tumour (lines: 67, 66c14, 168FARN, 4TO7, 68H, 64pT) as well as SP1 spontaneous CBA/J mouse mammary carcinoma. The difference was greatly reduced by neuraminidase treatment suggesting a masking of PNA binding sites by sialic acid. Separation of cell lysates by SDS-PAGE revealed a high molecular weight PNA binding glycoprotein (greater than 250 kd) expressed by normal mammary epithelium and preneoplastic D2HAN cells, but not by tumour cells regardless of neuraminidase treatment. A PNA reactive glycoprotein of approximately 90 kd was uniquely expressed in normal mammary epithelial lysates, although neuraminidase treatment exposed a similar band in a few tumour lines. Normal mammary epithelium, preneoplastic D2HAN cells, and the poorly tumorigenic clone D2.0R expressed a PNA binding glycoprotein of approximately 150 kd. This band appeared to be specifically sialylated during transition from the high PNA binding, low tumorigenic phenotype of D2.0R cells to the low PNA binding, highly tumorigenic phenotype of cells isolated from tumours resulting from s.c. implantation of D2.0R cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Glicoproteínas/metabolismo , Lectinas/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Mitogénicos/fisiología , Animales , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Femenino , Citometría de Flujo , Cinética , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Aglutinina de Mani , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Unión Proteica , Albúmina Sérica Bovina/metabolismo , Células Tumorales Cultivadas
14.
Neoplasma ; 39(3): 141-6, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1528317

RESUMEN

Bladder tumor cell lines derived from male F344 rats treated with N-buthyl N-(4-hydroxybuthyl) nitrosamine (BBN) or N-[4-(5-nitro-2-furyl)-2-thiazolyl] formamide (FANFT) have been established in vitro and characterized with respect to histology, karyotype, myc and c-Ha-ras oncogene expression or mutation, anchorage-independent growth and tumorigenicity in nude mice. This unique model system comprising 13 cell populations was employed to study common events during development of carcinogen-induced urothelial neoplasia. Differential expression of malignant phenotypes by these cell lines prompted us to examine their expression of carbohydrate structures binding peanut agglutinin (PNA), soy bean agglutinin (SBA) or leukoagglutinin (L-PHA), which are known indicators of tumor progression in rodents and humans. In the present study we analyzed the patterns of glycoproteins reactive with PNA and L-PHA by Western blotting. We also estimated quantitative differences in lectin binding to surfaces of normal rat urothelium and tumor cell lines by flow cytometry. The patterns of PNA or L-PHA reactive glycoproteins expressed by tumor cells were different from that of normal urothelium in culture. They were also different amongst the tumor cells. A unique non-sialylated, PNA binding glycoprotein (117 kD) was seen in the case of the highly tumorigenic F5 cell line and absent in normal urothelium as well as in other tumor cell lines. Normal cells did not express glycoprotein 60 kD binding PNA (only after desialylation), which was found in lysates of some but not all transformed cell lines. A very high molecular weight (much greater than 200), perhaps mucin-like sialoglycoprotein was found in normal urothelium but not in most of the tumor cell lines. Four major L-PHA reactive bands (greater than 200, 190, 100, 80 kD approximately) were found in normal urothelium. Some of those bands were overexpressed or missing in materials isolated from different tumor cell populations. Total cell surface binding of SBA and PNA by different tumor cell lines was very heterogenous (167-2% that of normal urothelium). No simple correlation between expression of the lectin binding glycoconjugates by urothelial carcinoma cells and other known functional, phenotypic or genetic alterations was found. We were also unable to demonstrate carcinogen-specific changes in expression of lectin binding to these tumor cell lines. Thus we conclude that lectin binding patterns are cell line specific. This may reflect distinct pathways of progression of individual cell lines. The potential sources of phenotypic variability between the cell lines were discussed.


Asunto(s)
Lectinas/metabolismo , Fitohemaglutininas/metabolismo , Lectinas de Plantas , Proteínas de Soja , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Sitios de Unión , Butilhidroxibutilnitrosamina , Aberraciones Cromosómicas , Electroforesis en Gel de Poliacrilamida , FANFT , Expresión Génica/genética , Genes myc/genética , Genes ras/genética , Glicoproteínas/análisis , Masculino , Ratones , Ratones Desnudos , Mutación , Proteínas de Neoplasias/análisis , Aglutinina de Mani , Ratas , Ratas Endogámicas F344 , Dodecil Sulfato de Sodio , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/genética
15.
Cancer Lett ; 57(1): 27-36, 1991 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2025877

RESUMEN

Fluorescent lectin binding to cell surfaces was quantitatively analysed by flow cytometry on mortal human breast epithelial cells MCF-10M, the immortalized cell line MCF-10A derived from MCF-10M and sublines of MCF-10A transfected with the neomycin resistance gene (MCF-10Aneo), the c-Ha-ras protooncogene (MCF-10AneoN), or transfected and transformed with the c-Ha-ras activated oncogene (MCF-10AneoT). Immortal MCF-10A cells bound 10-fold more peanut agglutinin (PNA) and soy bean agglutinin (SBA) than did MCF-10M cells. Transformed MCF-10AneoT cells bound approximately ten times more PNA than did non-transformed cells transfected with protooncogene (MCF-10AneoN). Treatment of the transfectants with neuraminidase abrogated the differences in PNA-binding and reduced the differences in SBA binding. SDS-PAGE separation of PNA binding glycoproteins revealed different patterns for all MCF-10A derived sublines.


Asunto(s)
Membrana Celular/metabolismo , Transformación Celular Neoplásica , Genes ras , Glicoproteínas de Membrana/metabolismo , Western Blotting , Mama , Línea Celular , Membrana Celular/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Epitelio/metabolismo , Femenino , Citometría de Flujo/métodos , Glicosilación , Humanos , Lectinas , Glicoproteínas de Membrana/aislamiento & purificación , Neuraminidasa/farmacología , Transfección
16.
Int J Cancer ; 47(3): 466-72, 1991 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-1993557

RESUMEN

Tumor subpopulations 66c14, 168FARN, and 4T07 are drug-resistant variants selected from sister subpopulations derived from a single mouse mammary tumor. These subpopulations are heterogeneous in their capacities to form experimental metastatic growth in the lungs and liver. Initial survival kinetics of arrested cells, determined by the clearance of 125IUdR-labelled cells, and subsequent growth rates, determined by sequential recovery of clonogenic tumor cells from occult metastases, both correlated with organ-colonizing potential as determined by necropsy. The growth rates of these 3 subpopulations were determined in vitro in monolayer and in situ in the subcutis, in the liver following intrasplenic injection, and in the lung following intravenous injection. Clonogenic potential of all 3 lines was similar in vitro (54-59%). Growth rates in vitro (population doubling times 16.5-21 hr) and in the subcutis (tumor volume doubling times 5.2-7.4 days) were similar for the 3 subpopulations, but differed significantly in the liver and lungs. For line 4T07, the most metastatic line to both lung and liver, population doubling times in vitro and in the lung and liver were similar, ranging from 17 to 26 hr. For lines 66c14 and 168FARN, the growth rates in lungs and livers were much slower than in vitro. Line 66c14, which is relatively more metastatic to the lungs, grew much faster in the lung (39 hours) than in the liver (91 hr), but line 168FARN, which is relatively more metastatic to the liver, grew at a faster rate in the liver (37 hr) than in the lung (63 hr). Thus, 3 tumor subpopulations (seeds) derived from a single tumor were differentially affected by host organ factors (soil) at 2 distinct stages in the metastatic process.


Asunto(s)
Neoplasias Hepáticas Experimentales/secundario , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia/patología , Animales , División Celular/fisiología , Supervivencia Celular/fisiología , Femenino , Idoxuridina/metabolismo , Inyecciones Intravenosas , Inyecciones Subcutáneas , Radioisótopos de Yodo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Bazo/patología , Factores de Tiempo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA