Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Cell Biol ; 178(3): 425-36, 2007 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-17664334

RESUMEN

Hypoxia activates genetic programs that facilitate cell survival; however, in cancer, it may promote invasion and metastasis. In this study, we show that breast cancer cells cultured in 1.0% O(2) demonstrate changes consistent with epithelial-mesenchymal transition (EMT). Snail translocates to the nucleus, and E-cadherin is lost from plasma membranes. Vimentin expression, cell migration, Matrigel invasion, and collagen remodeling are increased. Hypoxia-induced EMT is accompanied by increased expression of the urokinase-type plasminogen activator receptor (uPAR) and activation of cell signaling factors downstream of uPAR, including Akt and Rac1. Glycogen synthase kinase-3beta is phosphorylated, and Snail expression is increased. Hypoxia-induced EMT is blocked by uPAR gene silencing and mimicked by uPAR overexpression in normoxia. Antagonizing Rac1 or phosphatidylinositol 3-kinase also inhibits development of cellular properties associated with EMT in hypoxia. Breast cancer cells implanted on chick chorioallantoic membranes and treated with CoCl(2), to model hypoxia, demonstrate increased dissemination. We conclude that in hypoxia, uPAR activates diverse cell signaling pathways that cooperatively induce EMT and may promote cancer metastasis.


Asunto(s)
Neoplasias de la Mama , Epitelio/fisiología , Hipoxia , Mesodermo/fisiología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Animales , Antimutagênicos/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Cobalto/metabolismo , Colágeno/metabolismo , Activación Enzimática , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mesodermo/citología , Invasividad Neoplásica , Metástasis de la Neoplasia , Oxígeno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Proteína de Unión al GTP rac1/metabolismo
2.
J Neurosci Res ; 89(4): 544-51, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21290408

RESUMEN

Binding of activated α(2)-macroglobulin (α(2)M) to LDL receptor-related protein-1 (LRP1) in Schwann cells activates ERK/MAP kinase and Akt and thereby promotes cell survival and migration. The goal of this study was to determine whether α(2)M binding to LRP1 regulates expression of cytokines and chemokines. To assess the LRP1 response selectively, we studied primary cultures of rat Schwann cells. In a screening assay that detects 84 gene products, monocyte chemoattractant protein-1 (MCP-1/CCL2) mRNA expression was increased more than 13-fold in Schwann cells treated with activated α(2)M. The effects of α(2)M on MCP-1 expression were selective, because expression of the general proinflammatory cytokine tumor necrosis factor-α (TNF-α) was not induced. We confirmed that α(2)M selectively induces expression of MCP-1 and not TNF-α in single-target qPCR assays. MCP-1 protein accumulated at increased levels in conditioned medium of α(2)M-treated cells. LRP1 was necessary for induction of MCP-1 expression, as determined in experiments with the LRP1 antagonist receptor-associated protein, a mutated form of full-length α(2)M that does not bind LRP1, and in studies with Schwann cells in which LRP1 was silenced. Inhibiting ERK/MAP kinase activation blocked expression of MCP-1. These studies support a model in which LRP1 regulates multiple aspects of Schwann cell physiology in the response to PNS injury.


Asunto(s)
Quimiocina CCL2/biosíntesis , Regulación de la Expresión Génica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Células de Schwann/metabolismo , Transducción de Señal/fisiología , alfa-Macroglobulinas/metabolismo , Animales , Western Blotting , Citocinas/biosíntesis , Humanos , Ratones , Compresión Nerviosa , Reacción en Cadena de la Polimerasa , Unión Proteica , ARN Mensajero/análisis , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones
3.
J Proteome Res ; 9(12): 6689-95, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20919742

RESUMEN

LDL receptor-related protein 1 (LRP1) is an endocytic receptor, reported to regulate the abundance of other receptors in the plasma membrane, including uPAR and tissue factor. The goal of this study was to identify novel plasma membrane proteins, involved in cell-signaling, that are regulated by LRP1. Membrane protein ectodomains were prepared from RAW 264.7 cells in which LRP1 was silenced and control cells using protease K. Peptides were identified by LC-MS/MS. By analysis of spectral counts, 31 transmembrane and secreted proteins were regulated in abundance at least 2-fold when LRP1 was silenced. Validation studies confirmed that semaphorin4D (Sema4D), plexin domain-containing protein-1 (Plxdc1), and neuropilin-1 were more abundant in the membranes of LRP1 gene-silenced cells. Regulation of Plxdc1 by LRP1 was confirmed in CHO cells, as a second model system. Plxdc1 coimmunoprecipitated with LRP1 from extracts of RAW 264.7 cells and mouse liver. Although Sema4D did not coimmunoprecipitate with LRP1, the cell-surface level of Sema4D was increased by RAP, which binds to LRP1 and inhibits binding of other ligands. These studies identify Plxdc1, Sema4D, and neuropilin-1 as novel LRP1-regulated cell-signaling proteins. Overall, LRP1 emerges as a generalized regulator of the plasma membrane proteome.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/análisis , Proteómica/métodos , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células CHO , Línea Celular , Cromatografía Liquida , Cricetinae , Cricetulus , Immunoblotting , Inmunoprecipitación , Hígado/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Macrófagos/citología , Macrófagos/metabolismo , Espectrometría de Masas , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuropilina-1/metabolismo , Unión Proteica , Interferencia de ARN , Receptores de Superficie Celular/metabolismo , Receptores de LDL/genética , Semaforinas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética
4.
J Biol Chem ; 284(34): 22825-33, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19546228

RESUMEN

Hypoxia induces expression of the urokinase receptor (uPAR) and activates uPAR-dependent cell signaling in cancer cells. This process promotes epithelial-mesenchymal transition (EMT). uPAR overexpression in cancer cells also promotes EMT. In this study, we tested whether uPAR may be targeted to reverse cancer cell EMT. When MDA-MB 468 breast cancer cells were cultured in 1% O(2), uPAR expression increased, as anticipated. Cell-cell junctions were disrupted, vimentin expression increased, and E-cadherin was lost from cell surfaces, indicating EMT. Transferring these cells back to 21% O(2) decreased uPAR expression and reversed the signs of EMT. In uPAR-overexpressing MDA-MB 468 cells, EMT was reversed by silencing expression of endogenously produced urokinase-type plasminogen activator (uPA), which is necessary for uPAR-dependent cell signaling, or by targeting uPAR-activated cell signaling factors, including phosphatidylinositol 3-kinase, Src family kinases, and extracellular signal-regulated kinase. MDA-MB 231 breast cancer cells express high levels of uPA and uPAR and demonstrate mesenchymal cell morphology under normoxic culture conditions (21% O(2)). Silencing uPA expression in MDA-MB-231 cells decreased expression of vimentin and Snail, and induced changes in morphology characteristic of epithelial cells. These results demonstrate that uPAR-initiated cell signaling may be targeted to reverse EMT in cancer.


Asunto(s)
Epitelio/patología , Mesodermo/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Transducción de Señal , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Movimiento Celular , Embrión de Pollo , Pollos , Cromonas/farmacología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Flavonoides/farmacología , Humanos , Immunoblotting , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Microscopía Fluorescente , Morfolinas/farmacología , Oxígeno/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tubulina (Proteína)/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Vimentina/genética
5.
Am J Pathol ; 175(1): 190-200, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19497996

RESUMEN

The urokinase receptor (uPAR) promotes metastasis of human malignancies; however, its mechanism of action remains incompletely understood. Established models focus on the ability of uPAR to bind urokinase-type plasminogen activator (uPA) and promote protease activation in the tumor cell microenvironment; however, uPAR also regulates cell signaling and migration by both uPA-dependent and -independent mechanisms in vitro. The significance of uPAR as a cell-signaling receptor in vivo remains unclear. In this study, we expressed either human or mouse uPAR in human embryonic kidney (HEK-293) cells. We selected HEK-293 cells because, unlike most cancer cells, they do not express uPA or uPAR endogenously. Both mouse and human uPAR increased cell adhesion and migration on vitronectin. Rac1 was activated and responsible for the increase in cell migration. HEK-293 cells that did not express uPAR formed palpable tumors in severe combined immunodeficient mice; however, metastases were exceedingly rare. The xenografts contained abundant mouse uPA, produced by infiltrating mouse cells, but no human uPA. Mouse uPA bound only to mouse uPAR and not human uPAR and, thus, could not interact with human uPAR-expressing HEK-293 cells in xenografts. Nevertheless, both mouse and human uPAR significantly increased HEK-293 cell metastasis into the lungs. The activity of human uPAR suggests that uPAR may promote cancer metastasis independent of uPA. Candidate mechanisms include its effects on adhesion, migration, and Rac1 activation.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias Experimentales/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Western Blotting , Adhesión Celular/fisiología , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones SCID , Neoplasias Experimentales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo , Proteína de Unión al GTP rac1/metabolismo
6.
Cancer Res ; 65(21): 9829-33, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267005

RESUMEN

We have developed genetically fluorescent orthotopic models of human pancreatic cancer. In these models, noninvasive fluorescent protein imaging (FPI) of internal primary tumors and metastatic deposits has been carried out. Whole-body tumor images are easily and inexpensively obtained using FPI, permitting both detection and quantification of tumor load. In this study, we simultaneously compared single mice with a highly fluorescent, red fluorescent protein-expressing orthotopic pancreatic cancer xenografts with both FPI and high-resolution magnetic resonance imaging (MRI). Images were acquired at multiple time points after tumor implantation in the pancreas. Indwelling pancreatic primary tumors and metastatic foci were detected by both FPI and MRI. Moreover, a strong correlation existed between images taken with these two technologies. FPI permitted rapid, high-throughput imaging without the need for either anesthesia or contrast agents. Both FPI and MRI enabled accurate imaging of tumor growth and metastasis, although MRI enabled tissue structure to be visualized as well. FPI has high resolution and is exceedingly rapid with instant image capture. We suggest a complimentary role for these two imaging modalities.


Asunto(s)
Proteínas Luminiscentes/análisis , Neoplasias Pancreáticas/diagnóstico , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Trasplante Heterólogo , Proteína Fluorescente Roja
7.
Cancer Res ; 64(5): 1828-33, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14996746

RESUMEN

Adjuvant treatment with the cytosine analogue 1-(2-C-cyano-2-deoxy-beta-D-arabino-pentofuranosyl)-N(4)-palmitoylcytosine (CS-682) results in a highly significant increase in survival in the aggressive orthotopic MIA-PaCa-2 human pancreatic cancer mouse model. Seven days after implantation, mice were randomized into eight groups, depending on whether they were to be treated by tumor resection, 5 weeks of CS-682 chemotherapy at 40-60 mg/kg once daily, or both. Throughout the course of treatment, noninvasive optical whole-body imaging based on brilliant red fluorescent protein expression of the tumor permitted visualization and quantification of primary, metastatic, and recurrent disease. Total tumor burden negatively correlated with survival. Untreated mice died of disseminated disease with a median survival of 26 days. Surgical resection alone conferred a small but significant survival advantage (median survival, 28 days, P = 0.03). Primary CS-682 treatment at all doses also significantly prolonged survival compared with untreated animals (P < 0.05) and was more effective than surgery alone at doses of 50 and 60 mg/kg (median survival, 34 days, P = 0.045, and 38.5 days, P = 0.03, respectively). Maximal survival (median, 48 days, with 30% of animals surviving longer than 60 days) was achieved by adjuvant CS-682 (50 mg/kg), given after surgical resection of the primary pancreatic tumor (P = 0.004 compared with surgery alone). The results demonstrate that adjuvant oral administration of CS-682 for pancreatic cancer is highly effective with acceptable toxicity, suggesting its potential for cure of this disease in appropriate combinations.


Asunto(s)
Arabinonucleósidos/uso terapéutico , Citosina/análogos & derivados , Citosina/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Arabinonucleósidos/toxicidad , Línea Celular Tumoral , Quimioterapia Adyuvante , Citosina/toxicidad , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología
8.
Cancer Res ; 63(17): 5521-5, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500389

RESUMEN

In this study we demonstrate the ability of a novel, p.o.-administered cytosine analogue, CS-682, to effectively prolong survival and inhibit metastatic growth in an imageable orthotopic mouse model of pancreatic cancer. MIA-PaCa-2-RFP pancreatic cancer cells were transduced with the Discosoma red fluorescent protein (RFP) and orthotopically implanted onto the pancreas of nude mice. Tumor RFP fluorescence facilitated real-time, sequential imaging, and quantification of primary and metastatic growth and dissemination in vivo. Mice were treated with various p.o. doses of CS-682 on a five times per week schedule until death. At a dose of 40 mg/kg, CS-682 prolonged survival compared with untreated animals (median survival 35 days versus 17 days; P = 0.0008). At nontoxic doses, CS-682 effectively suppressed the rate of primary tumor growth. CS-682 also decreased the development of malignant ascites and the formation of metastases, which were reduced significantly in number in the diaphragm, lymph nodes, liver, and kidney. Selective RFP tumor fluorescence enabled noninvasive real-time comparison between groups during treatment and facilitated identification of micrometastases in solid organs at autopsy. Thus, we have demonstrated that CS-682 is an efficacious antimetastatic agent that significantly prolongs survival in an orthotopic model of pancreatic cancer. The antimetastatic efficacy of CS-682 and its p.o. availability confer significant advantages and clinical potential to this agent for pancreatic cancer.


Asunto(s)
Arabinonucleósidos/farmacología , Citosina/análogos & derivados , Citosina/farmacología , Proteínas Luminiscentes/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Arabinonucleósidos/efectos adversos , División Celular/efectos de los fármacos , División Celular/fisiología , Citosina/efectos adversos , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Desnudos , Microscopía Fluorescente , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción Genética , Células Tumorales Cultivadas , Pérdida de Peso/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Fluorescente Roja
9.
Clin Exp Metastasis ; 21(1): 7-12, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15065597

RESUMEN

In order to investigate the antitumor and antimetastatic efficacy of new chemotherapeutic agents, a novel, red-fluorescent, orthotopic model of pancreatic cancer was constructed in nude mice. MIA-PaCa-2 human pancreatic cancer cells were transduced with red fluorescent protein (RFP) and initially grown subcutaneously. Fluorescent tumor fragments were then transplanted onto the pancreas by surgical orthotopic implantation (SOI), facilitating high-resolution, real-time visualization of tumor and metastatic growth and dissemination in vivo. Tumor growth at the primary site was visible within the first postoperative week, while distant metastasis and the development of ascites became visible over the following week. This MIA-PaCa-2-RFP model produced extensive local disease and metastases to the retroperitoneum (100%), spleen (100%), intestinal and periportal lymph nodes (100%), liver (40%) and diaphragm (80%), and gave rise to malignant ascites and peritoneal carcinomatosis in 80% of cases. Growth and metastasis of tumor was more rapid and frequent than in previously described orthotopic pancreatic cancer models, leading to a median survival of only 21 days after tumor implantation. This unique, red fluorescent model rapidly and reliably simulates the highly aggressive course of human pancreatic cancer and can be easily non-invasively visualized in the live animal. The model can therefore be used for the discovery and evaluation of novel therapeutics for the treatment of this devastating disease.


Asunto(s)
Proteínas Luminiscentes/metabolismo , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/metabolismo , Tasa de Supervivencia , Proteína Fluorescente Roja
10.
Cell Signal ; 24(9): 1847-55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22617030

RESUMEN

Binding of urokinase-type plasminogen activator (uPA) to its receptor, uPAR, in estrogen receptor-α (ERα) expressing breast cancer cells, transiently activates ERK downstream of FAK, Src family kinases, and H-Ras. Herein, we show that when uPAR is over-expressed, in two separate ERα-positive breast cancer cell lines, ERK activation occurs autonomously of uPA and is sustained. Autonomous ERK activation by uPAR requires H-Ras and Rac1. A mutated form of uPAR, which does not bind vitronectin (uPAR-W32A), failed to induce autonomous ERK activation. Expression of human uPAR or mouse uPAR but not uPAR-W32A in MCF-7 cells provided a selection advantage when these cells were deprived of estrogen in cell culture for two weeks. Similarly, MCF-7 cells that express mouse uPAR formed xenografts in SCID mice that survived and increased in volume in the absence of estrogen supplementation, probably reflecting the pro-survival activity of phospho-ERK. Autonomous uPAR signaling to ERK was sensitive to the EGFR tyrosine kinase inhibitors, Erlotinib and Gefitinib. The transition in uPAR signaling from uPA-dependent and transient to autonomous and sustained is reminiscent of the transformation in ErbB2/HER2 signaling observed when this gene is amplified in breast cancer. uPAR over-expression may provide a pathway for escape of breast cancer cells from ERα-targeting therapeutics.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Clorhidrato de Erlotinib , Receptor alfa de Estrógeno/antagonistas & inhibidores , Estrógenos/deficiencia , Estrógenos/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Gefitinib , Humanos , Ratones , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
11.
J Cell Sci ; 122(Pt 8): 1155-62, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19299462

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease in which myelin is progressively degraded. Because degraded myelin may both initiate and accelerate disease progression, clearing degraded myelin from extracellular spaces may be critical. In this study, we prepared myelin vesicles (MV) from rat brains as a model of degraded myelin. Murine embryonic fibroblasts (MEFs) rapidly internalized MVs, which accumulated in lysosomes only when these cells expressed low-density lipoprotein receptor-related protein (LRP1). Receptor-associated protein (RAP), which binds LRP1 and inhibits interaction with other ligands, blocked MV uptake by LRP1-expressing MEFs. As a complementary approach, we prepared primary cultures of rat astrocytes, microglia and oligodendrocytes. All three cell types expressed LRP1 and mediated MV uptake, which was inhibited by RAP. LRP1 gene-silencing in oligodendrocytes also blocked MV uptake. Myelin basic protein (MBP), which was expressed as a recombinant protein, bound directly to LRP1. MBP-specific antibody inhibited MV uptake by oligodendrocytes. In experimental autoimmune encephalomyelitis in mice, LRP1 protein expression was substantially increased in the cerebellum and spinal cord. LRP1 colocalized with multiple CNS cell types. These studies establish LRP1 as a major receptor for phagocytosis of degraded myelin, which may function alone or in concert with co-receptors previously implicated in myelin phagocytosis.


Asunto(s)
Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Vaina de Mielina/metabolismo , Fagocitosis , Animales , Animales Recién Nacidos , Células Cultivadas , Cerebelo/metabolismo , Humanos , Ligandos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Lisosomas/metabolismo , Ratones , Proteína Básica de Mielina/metabolismo , Neuroglía/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de LDL/metabolismo , Médula Espinal/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rap/metabolismo
12.
J Surg Res ; 113(1): 151-60, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12943825

RESUMEN

BACKGROUND: Realistic models of pancreatic cancer are necessary to develop effective drugs for the disease. More aggressive tumor models enhanced by brighter fluorescent biomarkers to follow the disease in real time would enhance the ability to predict accurately the effect of novel therapeutics on this particularly malignant human cancer. MATERIALS AND METHODS: A novel, highly fluorescent, red fluorescent protein (RFP)-expressing pancreatic cancer model was orthotopically established in nude mice. The MIA-PaCa-2 human pancreatic cancer cell line was transduced with RFP and grown subcutaneously. Fluorescent tumor fragments were then surgically transplanted onto the nude mouse pancreas. Groups treated with intraperitoneal gemcitabine or intravenous irinotecan were sequentially imaged to compare, in real time, the antimetastatic and antitumor effects of these agents compared with untreated controls. RESULTS: Rapid tumor growth and widespread metastases developed in untreated mice within 2 weeks, leading to a median survival of 21 days. In contrast, significant tumor growth suppression and consequent increase in survival (32.5 days, P = 0.009) were achieved with CPT-11. Gemcitabine highly improved survival (72 days, P = 0.004) by inducing transient tumor regression over the first 3 weeks. However, at this time, growth and dissemination occurred despite continued treatment, suggesting the development of tumor resistance. The antimetastatic efficacy of each drug was followed noninvasively in real time by imaging the RFP-expressing tumor and metastases, and was confirmed by fluorescent open imaging of autopsy specimens. CONCLUSIONS: This highly metastatic model reliably simulates the aggressive course of human pancreatic cancer. Noninvasive, sequential imaging permits quantification of tumor growth and dissemination and, thereby, real time evaluation of therapeutic efficacy. These features make this model an ideal, preclinical system with which to study novel therapeutics for pancreatic cancer.


Asunto(s)
Camptotecina/análogos & derivados , Desoxicitidina/análogos & derivados , Proteínas Luminiscentes/genética , Modelos Animales , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Camptotecina/uso terapéutico , Desoxicitidina/uso terapéutico , Humanos , Irinotecán , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/diagnóstico , Metástasis de la Neoplasia/tratamiento farmacológico , Valor Predictivo de las Pruebas , Transducción Genética , Células Tumorales Cultivadas , Gemcitabina , Proteína Fluorescente Roja
13.
Ann Surg Oncol ; 10(7): 762-72, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12900367

RESUMEN

BACKGROUND: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in malignant cells but not in normal cells. Ad/g-TRAIL, an adenoviral vector in which expression of green fluorescent protein (GFP) and TRAIL is driven by a human telomerase reverse transcriptase promoter, has shown promise as a targeted antitumor agent. METHODS: To investigate the effects of TRAIL gene therapy on pancreatic cancer, BxPC-3, MIA-PaCa-2, Panc-1, and ASPC-1 cells were treated with Ad/g-TRAIL. Transfection and protein expression were determined by using immunoblotting and identification of GFP with fluorescent microscopy and flow cytometry. Cell viability was determined by proliferation assay. Cell-cycle analysis and quantification of caspase-3 were used to identify apoptosis. The in vivo efficacy of Ad/g-TRAIL was characterized in a novel red fluorescent protein murine model of MIA-PaCa-2 pancreatic cancer. RESULTS: Cells treated with Ad/g-TRAIL expressed GFP and exhibited apoptotic morphology within 2 days of treatment. Treatment with this vector in vitro resulted in less cell viability, increased caspase-3 activity, and a greater apoptotic fraction than treatment with controls. In vivo, treatment with Ad/g-TRAIL significantly suppressed tumor growth. CONCLUSIONS: TRAIL gene therapy induces apoptosis of pancreatic tumor cells both in vitro and in vivo and is a promising therapy in the treatment of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Terapia Genética , Proteínas Luminiscentes/uso terapéutico , Glicoproteínas de Membrana/uso terapéutico , Neoplasias Pancreáticas/terapia , Telomerasa/uso terapéutico , Factor de Necrosis Tumoral alfa/uso terapéutico , Adenoviridae , Animales , Proteínas Reguladoras de la Apoptosis , Western Blotting , Caspasa 3 , Caspasas/metabolismo , Dominio Catalítico , Proteínas de Unión al ADN , Genes Transgénicos Suicidas , Proteínas Fluorescentes Verdes , Humanos , Insectos Vectores , Proteínas Luminiscentes/metabolismo , Ratones , Ligando Inductor de Apoptosis Relacionado con TNF , Transfección , Células Tumorales Cultivadas , Proteína Fluorescente Roja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA