Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Langmuir ; 34(3): 1178-1189, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-28976765

RESUMEN

Polyelectrolyte multilayers (PEMs) are of great interest as cell culture surfaces because of their ability to modify topography and surface energy and release biologically relevant molecules such as growth factors. In this work, fibroblast growth factor 2 (FGF2) was adsorbed directly onto polystyrene, plasma-treated polystyrene, and glass surfaces with a poly(methacrylic acid) and poly-l-histidine PEM assembled above it. Up to 14 ng/cm2 of FGF2 could be released from plasma-treated polystyrene surfaces over the course of 7 days with an FGF2 solution concentration of 100 µg/mL applied during the adsorption process. This release rate could be modulated by adjusting the adsorption concentration, decreasing to as low as 2 ng/cm2 total release over 7 days using a 12.5 µg/mL FGF2 solution. The surface energy and roughness could also be regulated using the adsorbed PEM. These properties were found to be substrate- and first-layer-dependent, supporting current theories of PEM assembly. When released, FGF2 from the PEMs was found to significantly enhance fibroblast proliferation as compared to culture conditions without FGF2. The results showed that growth factor release profiles and surface properties are easily controllable through modification of the PEM assembly steps and that these strategies can be effectively applied to common cell culture surfaces to control the cell fate.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/farmacología , Polielectrolitos/química , Adsorción , Proliferación Celular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Propiedades de Superficie
2.
Mol Carcinog ; 52 Suppl 1: E80-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23390063

RESUMEN

A single-nucleotide polymorphism (rs2274223: A5780G:His1927Arg) in the phospholipase C epsilon gene (PLCϵ) was recently identified as a susceptibility locus for esophageal cancer in Chinese subjects. To determine the underlying mechanisms of PLCϵ and this SNP in esophageal carcinogenesis, we analyzed PLCϵ genotypes, expression, and their correlation in esophageal cancer cell lines, non-transformed esophageal cells, 58 esophageal squamous cell carcinomas and 10,614 non-cancer subjects from China. We found that the G allele (AG or GG) was associated with increased PLCϵ mRNA and protein expression in esophageal cancer tissues and in esophageal cancer cell lines. G allele was also associated with higher enzyme activity, which might be associated with increased protein expression. Quantitative analysis of the C2 domain sequences revealed that A:G allelic imbalance was strongly linked to esophageal malignancy. Moreover, the analysis of 10,614 non-cancer subjects demonstrated that the G allele was strongly associated with moderate to severe esophagitis in the subjects from the high-incidence areas of China (OR 6.03, 95% CI 1.59-22.9 in high-incidence area vs. OR 0.74, 95% CI 0.33-1.64 in low-incidence area; P = 0.008). In conclusion, the PLCϵ gene, particularly the 5780G allele, might play a pivotal role in esophageal carcinogenesis via upregulating PLCϵ mRNA, protein, and enzyme activity, and augmenting inflammatory process in esophageal epithelium. Thus, 5780G allele may constitute a promising biomarker for esophageal squamous cell carcinoma risk stratification, early detection, and progression prediction.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Esofagitis/genética , Fosfoinositido Fosfolipasa C/genética , Polimorfismo de Nucleótido Simple/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Estudios de Casos y Controles , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/patología , Esofagitis/enzimología , Esofagitis/patología , Genotipo , Humanos , Immunoblotting , Técnicas para Inmunoenzimas , Estadificación de Neoplasias , Fosfoinositido Fosfolipasa C/metabolismo , Reacción en Cadena de la Polimerasa , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Tumorales Cultivadas
3.
Carcinogenesis ; 32(7): 986-94, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21470957

RESUMEN

The human olfactomedin 4 gene (OLFM4) encodes an olfactomedin-related glycoprotein. OLFM4 is normally expressed in a limited number of tissues, including the prostate, but its biological functions in prostate are largely unknown. In this study, we found that OLFM4 messenger RNA was reduced or undetectable in prostate cancer tissues and prostate cancer cell lines. To study the effects of OLFM4 on prostate cancer progression, we transfected PC-3 prostate cancer cells with OLFM4 to establish OLFM4-expressing PC-3 cell clones. The OLFM4-expressing PC-3 cell clones were found to have decreased proliferation and invasiveness compared with vector-transfected control PC-3 cells in vitro. In addition, nude mice injected with OLFM4-expressing PC-3 cells demonstrated reduced tumor growth and bone invasion and metastasis compared with mice injected with vector-transfected control cells. Mechanistic studies revealed that OLFM4 may exhibit its anticancer effects through regulating cell autophagy by targeting cathepsin D, as OLFM4 reduced cathepsin D protein levels and enzymatic activity and attenuated cathepsin D-induced cancer cell proliferation. In addition, overexpression of OLFM4 abrogated stromal cell derived factor-1 (SDF-1)-induced PC-3 cell invasiveness in a Matrigel invasion assay, partially through blocking SDF-1-mediated AKT phosphorylation. Coimmunoprecipitation and immunofluorescence staining studies in OLFM4-expressing PC-3 cells demonstrated a direct interaction between OLFM4 and cathepsin D or SDF-1. Taken together, these results suggest that OLFM4 negatively interacts with cathepsin D and SDF-1 and inhibits prostate cancer growth and bone metastasis.


Asunto(s)
Catepsina D/metabolismo , División Celular , Quimiocina CXCL12/metabolismo , Factor Estimulante de Colonias de Granulocitos/fisiología , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Humanos , Masculino , Neoplasias de la Próstata/metabolismo
4.
Sci Rep ; 11(1): 9808, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33963247

RESUMEN

Growth factor-eluting polymer systems have been widely reported to improve cell and tissue outcomes; however, measurements of actual growth factor concentration in cell culture conditions are limited. The problem is compounded by a lack of knowledge of growth factor half-lives, which impedes efforts to determine real-time growth factor concentrations. In this work, the half-life of basic fibroblast growth factor (FGF2) was determined using enzyme linked immunosorbent assay (ELISA). FGF2 release from polyelectrolyte multilayers (PEMs) was measured and the data was fit to a simple degradation model, allowing for the determination of FGF2 concentrations between 2 and 4 days of culture time. After the first hour, the FGF2 concentration for PEMs assembled at pH = 4 ranged from 2.67 ng/mL to 5.76 ng/mL, while for PEMs assembled at pH = 5, the concentration ranged from 0.62 ng/mL to 2.12 ng/mL. CRL-2352 fibroblasts were cultured on PEMs assembled at pH = 4 and pH = 5. After 2 days, the FGF2-eluting PEM conditions showed improved cell count and spreading. After 4 days, only the pH = 4 assembly condition had higher cells counts, while the PEM assembled at pH = 5 and PEM with no FGF2 showed increased spreading. Overall, the half-life model and cell culture study provide optimal concentration ranges for fibroblast proliferation and a framework for understanding how temporal FGF2 concentration may affect other cell types.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Fibroblastos/metabolismo , Polielectrolitos/química , Línea Celular , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Semivida , Humanos
5.
J Colloid Interface Sci ; 570: 135-142, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32146240

RESUMEN

Polyelectrolyte multilayers (PEMs) are a versatile category of materials due to their ability to modify surface properties for applications ranging from protective coatings to improved cell adhesion. Polyelectrolyte choice, including its structure and molecular weight (MW), is known to greatly influence PEM assembly and surface properties. In this work, poly(acrylic acid)/poly-l-lysine PEMs using three pairs of MWs (1.8k/15-30k, 100k/120k, and 250k/275k) were studied to determine the effects of their MWs on PEM assembly, topography and surface energy. PEMs assembly was monitored in a quartz crystal microbalance with dissipation, resulting in masses of 3.90 ± 0.87 µg/cm2, 10.80 ± 4.189 µg/cm2, and 30.04 ± 13.68 µg/cm2 for 10 bilayers of low, medium, and high MW pairs, respectively. The low MW PEM was more rigid. Low and high MW PEMs exhibited higher roughness than medium MW, caused by polyelectrolyte stripping. Surface energy remained constant with bilayer count in the low and high MW PEMs, but steadily increased in the medium MW PEM. Differences between medium MW PEMs from low and high MW systems indicate that, while PEM properties change with MW, they are not monotonically correlated and are instead related to changes in internal charge distributions and the resultant stripping that may occur.

6.
Clin Cancer Res ; 14(4): 1041-9, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18281536

RESUMEN

PURPOSE: hGC-1 (human granulocyte colony-stimulating factor-stimulated clone 1) is a gastrointestinal protein that is a member of the olfactomedin glycoprotein family. Its biological function remains poorly understood. Aberrant expression of hGC-1 in some human carcinomas has been recently reported. The purpose of this study was to examine hGC-1 expression in colon carcinoma and explore the relationship between hGC-1 expression and the clinicopathologic features of patients with colon cancer. EXPERIMENTAL DESIGN: The expression of hGC-1 in colon adenocarcinoma tissues was examined by dot-blot analysis, in situ hybridization, and immunohistochemistry. The association of hGC-1 expression pattern with patient differentiation grade, tumor stage, metastasis, and survival were examined. To further investigate the involvement of hGC-1 in colon cancer progression, human colon carcinoma (HT-29) cells overexpressing hGC-1 were established and cell proliferation, adhesion, and migration were studied. RESULTS: Compared with normal colon mucosa, the up-regulation of hGC-1 was more frequently detected in more differentiated colon cancers, whereas down-regulation or no expression was associated with poorly differentiated colon cancers. Interestingly, hGC-1 down-regulation was also found in late tumor-node-metastasis stage, metastasis, and in patients with shorter survival. The morphology and cortical actin distribution of HT-29 cells were altered by hGC-1 overexpression. However, this did not change cell proliferation, but decreased cell adhesion and migration. CONCLUSION: Our findings indicate that hGC-1 is involved in colon cancer adhesion and metastasis, and that hGC-1 may be a useful marker for tumor differentiation and progression of human colon carcinoma.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Adenocarcinoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias del Colon/genética , Metilación de ADN , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Células HT29 , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Estimación de Kaplan-Meier , Masculino , Microscopía Confocal , Persona de Mediana Edad , Regiones Promotoras Genéticas , ARN Mensajero/análisis
7.
Colloids Surf B Biointerfaces ; 178: 276-284, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30878802

RESUMEN

Polyelectrolyte multilayers (PEMs) are a category of materials commonly used as coatings on surfaces that interact with cells. The properties of PEMs have been established to be controlled by not only polyelectrolyte choice, but by the identity of the initially applied (bottom) layer. In this work, 5-bilayer PEMs consisting of poly(diallyldimethylammonium chloride) (PDADMAC) and poly(sodium 4-styrenesulfonate) (PSS) were coated on gold-sputtered quartz substrates with different first layer materials. A final poly-l-lysine (PLL) layer was added to all PEMs to provide identical top layers conducive to cell growth. As in previous work, initial layer selection affected PEM roughness. All coated surfaces, including the PLL-only control, showed increased dispersive surface energy but decreased polar surface energy, as compared to uncoated surfaces. When human mesenchymal stem cells (hMSCs) were cultured on these surfaces, analysis through lateral cell imaging for the first 90 min and fluorescent staining after 1 day showed improved attachment on surfaces with a PDADMAC bottom layer. However, after 4 days, a higher cell density was observed on the PLL-only and uncoated control surfaces, indicating that the PEMs negatively affected hMSC proliferation. Both the long and short time period results did not correlate to any of the roughness and surface energy trends, indicating more complex interactions between the cells and the surface relating to charge distribution and functional group density.


Asunto(s)
Células Madre Mesenquimatosas/citología , Polielectrolitos/química , Polilisina/química , Línea Celular , Proliferación Celular/fisiología , Humanos
8.
Clin Cancer Res ; 13(16): 4891-9, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699869

RESUMEN

PURPOSE: The lack of effective treatment for pancreatic cancer results in a very low survival rate. This study explores the enhancement of the therapeutic effect on human pancreatic cancer via the combination of triptolide and ionizing radiation (IR). EXPERIMENTAL DESIGN: In vitro AsPC-1 human pancreatic cancer cells were treated with triptolide alone, IR alone, or triptolide plus IR. Cell proliferation was analyzed with sulforhodamine B (SRB) method and clonogenic survival; comparison of apoptosis induced by the above treatment was analyzed by annexin V-propidium iodide (PI) staining. Furthermore, the expression of apoptotic pathway intermediates was measured by the assay of caspase activity and Western blot. Mitochondrial transmembrane potential was determined by JC-1 assay. In vivo, AsPC-1 xenografts were treated with 0.25 mg/kg triptolide, 10 Gy IR, or triptolide plus IR. The tumors were measured for volume and weight at the end of the experiment. Tumor tissues were tested for terminal nucleotidyl transferase-mediated nick end labeling (TUNEL) and immunohistochemistry. RESULTS: The combination of triptolide plus IR reduced cell survival to 21% and enhanced apoptosis, compared with single treatment. In vivo, tumor growth of AsPC-1 xenografts was reduced further in the group treated with triptolide plus IR compared with single treatment. TUNEL and immunohistochemistry of caspase-3 cleavage in tumor tissues indicated that the combination of triptolide plus IR resulted in significantly enhanced apoptosis compared with single treatments. CONCLUSIONS: Triptolide in combination with ionizing radiation produced synergistic antitumor effects on pancreatic cancer both in vitro and in vivo and seems promising in the combined modality therapy of pancreatic cancer.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Diterpenos/farmacología , Neoplasias Pancreáticas/terapia , Fenantrenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Terapia Combinada , Compuestos Epoxi/farmacología , Fase G2/efectos de los fármacos , Fase G2/efectos de la radiación , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/radioterapia , Trasplante Heterólogo
9.
Adv Exp Med Biol ; 599: 109-16, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17727254

RESUMEN

The biological and physiological effects of exogenous FGF 1 and VEGF were measured using the KHT murine fibrosarcoma tumor model. Tumor-bearing C3H mice were treated intratumorally with either one or six daily doses of 6 microg/mouse FGF1, VEGF, or saline. Tumors were excised 24 hrs after the final injection. Compared to controls, only FGF1 treatment significantly increased tumor weight and size, and only in the 6 dose group. Both FGF1 and VEGF administration (6 dose) decreased tumor cell hypoxia as detected by EF5 uptake: 85% +/- 5% for FGF1 and 82% +/- 6% for VEGF versus 100% +/- 6% for controls. Decreased tumor cell EF5 staining, however, was not associated with changes in numbers of structural or angiogenic vessels. DiOC7 staining showed a slight decrease in perfused vessel numbers in tumors treated with daily VEGF. Intratumoral injections of FGF1 or VEGF also slightly decreased the tumor tissue chemokine MCP-1, interleukins (IL-1beta, IL-6, and IL-18) mRNA expression, and increased NFkappaB binding without altering Ap-1 binding of IkappaB protein expression. In summary, single pulse exposures of tumors to angiogenic factors had little or no effects on tumor growth or perfusion, while daily exposures stimulated tumor growth through improved tumor oxygenation. This improved vascular function occurs without an increase in vascular density.


Asunto(s)
Quimiocina CCL2/metabolismo , Factor 1 de Crecimiento de Fibroblastos/uso terapéutico , Interleucinas/metabolismo , Neoplasias Experimentales , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Femenino , Factor 1 de Crecimiento de Fibroblastos/administración & dosificación , Humanos , Interleucinas/genética , Ratones , Ratones Endogámicos C3H , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
10.
Adv Exp Med Biol ; 599: 147-53, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17727259

RESUMEN

Endostatin, a fragment of the C-terminal domain of mouse collagen XVIII, is a recently demonstrated endogenous inhibitor of tumor angiogenesis. Although endostatin can be detected in blood and urine of tumor-bearing as well as normal mice, the exact localization of the endogenous protein and its related peptides in tumor tissues is unknown. We used immunohistochemistry and immunoblotting to identify endostatin tissue location and staining patterns in tumor, as well as to determine the differences in the levels of endostatin expression between tumor cells (in vitro) and tumor tissues (in vivo). Using a specific polyclonal antibody against murine endostatin, we quantitatively determined the levels of endostatin in five murine mammary tumors and the KHT sarcoma by Western blotting. The staining patterns for this protein in tumor sections were examined histologically by immunohistochemistry. Our results show that: (1) Endogenous endostatin and its related peptides are widely distributed in all in vivo tumor types tested, but not in most of the cultured tumor cell lines. (2) Endogenous endostatin stained most tumor stromal components, including vessel walls, basement membranes, extracellular spaces, and tumor cells. (3) Staining patterns and localization of endostatin and thrombospondin-1 were similar in these tumor sections.


Asunto(s)
Inhibidores de la Angiogénesis/análisis , Endostatinas/análisis , Neoplasias/química , Péptidos/análisis , Animales , Línea Celular Tumoral/química , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C3H , Neoplasias/patología , Trombospondinas/análisis
11.
Int J Radiat Oncol Biol Phys ; 65(3): 890-8, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16751071

RESUMEN

PURPOSE: To determine whether curcumin ameliorates acute and chronic radiation skin toxicity and to examine the expression of inflammatory cytokines (interleukin [IL]-1, IL-6, IL-18, IL-1Ra, tumor necrosis factor [TNF]-alpha, and lymphotoxin-beta) or fibrogenic cytokines (transforming growth factor [TGF]-beta) during the same acute and chronic phases. METHODS AND MATERIALS: Curcumin was given intragastrically or intraperitoneally to C3H/HeN mice either: 5 days before radiation; 5 days after radiation; or both 5 days before and 5 days after radiation. The cutaneous damage was assessed at 15-21 days (acute) and 90 days (chronic) after a single 50 Gy radiation dose was given to the hind leg. Skin and muscle tissues were collected for measurement of cytokine mRNA. RESULTS: Curcumin, administered before or after radiation, markedly reduced acute and chronic skin toxicity in mice (p < 0.05). Additionally, curcumin significantly decreased mRNA expression of early responding cytokines (IL-1 IL-6, IL-18, TNF-alpha, and lymphotoxin-beta) and the fibrogenic cytokine, TGF-beta, in cutaneous tissues at 21 days postradiation. CONCLUSION: Curcumin has a protective effect on radiation-induced cutaneous damage in mice, which is characterized by a downregulation of both inflammatory and fibrogenic cytokines in irradiated skin and muscle, particularly in the early phase after radiation. These results may provide the molecular basis for the application of curcumin in clinical radiation therapy.


Asunto(s)
Curcumina/uso terapéutico , Citocinas/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Piel/efectos de la radiación , Animales , Evaluación Preclínica de Medicamentos/métodos , Femenino , Interleucina-1/metabolismo , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Linfotoxina-alfa/metabolismo , Linfotoxina beta , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , ARN Mensajero/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Piel/metabolismo
12.
Radiat Res ; 165(2): 181-91, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16435917

RESUMEN

Interleukin 1 beta (IL1B), a potent pro-inflammatory cytokine, is directly up-regulated by radiation and is known to regulate other inflammation-related molecules, such as the matrix metalloproteinases (MMPs) and their endogenous inhibitors (TIMPs). However, the nature of the interaction of IL1B with MMPs and TIMPs in radiation-induced skin fibrosis is unknown. We examined the response of primary dermal keratinocytes, fibroblasts and endothelial cells to single-fraction radiation (10 Gy) and compared the results to a temporal sequence of histology from irradiated C57BL/6 and IL1R1 knockout mice. These studies showed that keratinocytes are the major IL1-producing cells in vitro and that radiation induces an immediate and chronic elevation in the expression of IL1B mRNA in the skin of C57BL/6 mice. This elevation was principally early and was less pronounced in the IL1R1 knockout strain, which also demonstrated reduced late radiation fibrosis. Radiation also increased expression of MMP mRNA in C57BL/6 mice. Finally, exogenous IL1B protein induced robust endogenous IL1B mRNA expression, along with a brisk increase in MMPs and collagen III, but only in the C57BL/6 mice. In conclusion, these data suggest that IL1B plays a critical role in radiation-induced fibrosis and that the increased MMPs fail to block the IL1-related collagen accumulation.


Asunto(s)
Interleucina-1/metabolismo , Radiodermatitis/metabolismo , Transducción de Señal/efectos de la radiación , Piel/metabolismo , Piel/efectos de la radiación , Animales , Femenino , Fibrosis/metabolismo , Rayos gamma/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Piel/patología
13.
Cancer Res ; 64(16): 5712-9, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15313911

RESUMEN

Although clinical trials of antiangiogenic strategies have been disappointing when administered as single agents, such approaches can play an important role in cancer treatment when combined with conventional therapies. Previous studies have shown that DC101, an antiangiogenic monoclonal antibody against vascular endothelial growth factor receptor-2, can produce significant growth inhibition in spontaneous and transplanted tumors but can also induce substantial hypoxia. Because DC101 appears to potentiate radiotherapy in some tumors, the present studies were undertaken to characterize pathophysiological changes following combined therapy and to determine whether radioresponse is enhanced despite the induction of hypoxia. MCa-4 and MCa-35 mammary carcinomas were treated with: (a) DC101; (b) 5 x 6 Gy radiation fractions; or (c) the combination. Image analysis of frozen tumor sections was used to quantitate: (a) hypoxia; (b) spacing of total and perfused blood vessels; and (c) endothelial and tumor cell apoptosis. For MCa-4, combination treatment schedules produced significant and prolonged delays in tumor growth, whereas single-modality treatments had minor effects. For MCa-35, radiation or the combination led to equivalent growth inhibition. In all tumors, hypoxia increased markedly after either radiation or DC101 alone. Although combination therapy produced no immediate pathophysiological changes, hypoxia ultimately increased after cessation of therapy. Preferential increases in endothelial apoptosis following combination treatment suggest that in addition to blocking tumor angiogenesis, DC101 enhances radiotherapy by specifically sensitizing endothelial cells, leading to degeneration of newly formed blood vessels.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Mamarias Experimentales/radioterapia , Neoplasias Mamarias Experimentales/terapia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , División Celular/fisiología , División Celular/efectos de la radiación , Hipoxia de la Célula/fisiología , Hipoxia de la Célula/efectos de la radiación , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Endotelio Vascular/patología , Endotelio Vascular/efectos de la radiación , Femenino , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos C3H , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
14.
Cancer Res ; 63(15): 4684-91, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12907650

RESUMEN

Alteration of the phenotype of breast cancers from estrogen-dependent to estrogen-independent growth often leads to the failure of antiestrogenic tumor therapies. We report that overexpression of vascular endothelial growth factor (VEGF) by estrogen-dependent MCF-7 breast cancer cells could abolish estrogen-dependent tumor growth in ovariectomized mice. In the absence of estrogen, MCF-7 VEGF-expressing tumors with increased vessel density showed growth kinetics similar to, or even greater than, that of parental MCF-7 tumors with estrogen supplementation. Overexpression of VEGF by MCF-7 cells or treatment on parental MCF-7 cells with recombinant VEGF also stimulated cell proliferation in culture. Our data suggest that VEGF stimulation of MCF-7 tumor angiogenesis and growth is mediated by both autocrine and paracrine mechanisms.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factores de Crecimiento Endotelial/biosíntesis , Estradiol/farmacología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Linfocinas/biosíntesis , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , División Celular/efectos de los fármacos , División Celular/fisiología , Factores de Crecimiento Endotelial/genética , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Operón Lac , Linfocinas/genética , Ratones , Ratones Desnudos , Neoplasias Hormono-Dependientes/irrigación sanguínea , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Isoformas de Proteínas , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Transfección , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
15.
J Clin Oncol ; 22(11): 2207-13, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15169810

RESUMEN

PURPOSE: Fibrotic sequelae remain the most important dose-limiting toxicity of radiation therapy to soft tissue. Functionally, this is reflected in loss of range of motion and muscle strength and the development of limb edema and pain. Tumor necrosis factor alpha and fibroblast growth factor 2 (FGF2), which are abnormally elevated in irradiated tissues, may mediate radiation fibrovascular injury. PATIENTS AND METHODS: In an open label drug trial, we studied the effects of pentoxifylline (400 mg orally tid for 8 weeks) on 30 patients who displayed late, radiation-induced fibrosis at 1 to 29 years posttreatment (40 to 84 Gy). The primary outcome measurement was change in physical impairments thought to be secondary to radiation, including active and passive range of motion (AROM and PROM), muscle strength, limb edema, and pain. Plasma levels of cytokines (tumor necrosis factor alpha and FGF2) also were measured. Twenty-seven patients completed baseline and 8-week assessments, and 24 patients completed baseline, 8-week, and 16-week assessments. RESULTS: After 8 weeks of pentoxifylline intervention, 20 of 23 patients with impaired AROM and 19 of 22 with impaired PROM improved; 11 of 19 patients with muscle weakness showed improved motor strength; five of seven patients with edema had decreased limb girth; and nine of 20 patients had decreased pain. Pretreatment FGF2 levels dropped from an average of 44.9 pg/mL to 24.0 pg/mL after 8 weeks of treatment. CONCLUSION: Patients receiving pentoxifylline demonstrated improved AROM, PROM, and muscle strength and decreased limb edema and pain. Reversal of these delayed radiation effects was associated with a decrease in circulating FGF2.


Asunto(s)
Pentoxifilina/uso terapéutico , Traumatismos por Radiación/tratamiento farmacológico , Protectores contra Radiación/uso terapéutico , Radioterapia/efectos adversos , Traumatismos de los Tejidos Blandos/tratamiento farmacológico , Adolescente , Adulto , Anciano , Citocinas/sangre , Citocinas/efectos de los fármacos , Fibrosis , Humanos , Persona de Mediana Edad , Pentoxifilina/farmacología , Traumatismos por Radiación/etiología , Protectores contra Radiación/farmacología , Rango del Movimiento Articular/efectos de los fármacos , Traumatismos de los Tejidos Blandos/etiología , Resultado del Tratamiento
16.
Int J Radiat Oncol Biol Phys ; 62(1): 273-8, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15850933

RESUMEN

PURPOSE: The negative effects of radiation on the bowel critically limit the treatment doses possible for tumors in the abdomen. The purpose of the present study was to measure mRNA levels of inflammatory cytokines in abdominally irradiated mouse bowel. METHODS AND MATERIALS: Eight- to 12-week-old DBA mice were irradiated to the whole bowel in single fractions of 0 (mock irradiation), 12.5, or 13.5 Gy, and sacrificed 18-25 weeks thereafter. Gross bowel reactions were scored for bowel retraction, bowel wall thickening, mesenteric telangiectasia, and petechia. Tissues were snap frozen and processed for RNase protection assay or reverse transcription polymerase chain reaction assay, or both. Transforming growth factor beta1 (TGFbeta1), TGFbeta2, TGFbeta3, tumor necrosis factor alpha, interleukin-6, and interferon gamma mRNA were measured. RESULTS: Radiation at 12.5 Gy and at 13.5 Gy produced significant bowel damage. Levels of all cytokines in irradiated mice were significantly increased (p < 0.05). CONCLUSIONS: Late radiation-related bowel fibrovascular toxicity includes cytokine signal pathways that parallel those of many other normal tissues. These cytokine responses include elevations of tumor necrosis factor alpha, TGFbeta1, and interleukin-6. There exist approaches for lowering these cytokine levels that do not also protect tumor, and thus a therapeutic gain is expected. Opportunities to use these cytokine measurements both to predict clinical toxicity and to develop interventions are discussed.


Asunto(s)
Citocinas/metabolismo , Intestinos/efectos de la radiación , Traumatismos Experimentales por Radiación/metabolismo , Animales , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos DBA , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta2 , Factor de Crecimiento Transformador beta3 , Factor de Necrosis Tumoral alfa/metabolismo
17.
Am J Clin Pathol ; 124(2): 282-7, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16040301

RESUMEN

Overexpression of decoy receptor (DcR) 3 protein, a recently discovered member of the tumor necrosis factor receptor superfamily, was examined in 40 esophagogastrectomy specimens containing areas of Barrett esophagus (n = 27), low-grade dysplasia (n = 27), high-grade dysplasia or carcinoma in situ (n = 22), and esophageal adenocarcinoma (EAC; n = 28) with immunohistochemical analysis. The results revealed significantly more overexpression of DcR3 in high-grade dysplasia or carcinoma in situ and EAC than in benign esophageal mucosa (both P < .0001), Barrett esophagus (both P < .001), and low-grade dysplasia (P < .01 and P = .033, respectively). Low-grade dysplasia also showed significant overexpression of DcR3 compared with benign esophagus (P < .05) but not with Barrett esophagus (P > .05). DcR3 overexpression seems to negatively correlate with the grade of EAC. Our results suggest that overexpression of DcR3 protein might aid in the diagnosis of high-grade dysplasia or carcinoma in situ and EAC and also might serve as a potential therapeutic target.


Asunto(s)
Adenocarcinoma/metabolismo , Biomarcadores de Tumor/análisis , Neoplasias Esofágicas/metabolismo , Glicoproteínas de Membrana/biosíntesis , Lesiones Precancerosas/metabolismo , Receptores de Superficie Celular/biosíntesis , Receptores del Factor de Necrosis Tumoral/biosíntesis , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Esofágicas/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/patología , Miembro 6b de Receptores del Factor de Necrosis Tumoral , Regulación hacia Arriba
18.
Clin Cancer Res ; 9(2): 562-70, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12576419

RESUMEN

BACKGROUND: Flavopiridol is a flavonoid with antiproliferative effects mediated, in part, by inhibition of cyclin-dependent kinases. Clinical manifestations in a previous Phase I trial in patients with refractory malignancies treated with a 72-h flavopiridol infusion included a proinflammatory syndrome consisting of fever, fatigue, and "local" tumor pain with concomitant alterations in plasma acute-phase reactant proteins. PURPOSE: The aim of this study was to determine whether the proinflammatory syndrome observed in this trial was associated with modulation of plasma cytokines. METHODS: Patients receiving flavopiridol (n = 76) had serial plasma samples drawn preinfusion and during the infusion for evaluation of interleukin (IL)-6, IL-10, IL-12, granulocyte macrophage colony-stimulating factor, basic-fibroblast growth factor, transforming growth factor-beta, and tumor necrosis factor-alpha levels by standard ELISA assays. The Wilcoxon signed rank test was used to test the significance of the difference between the baseline (time 0) plasma cytokine levels compared with the values of each subsequent data collection time points (8, 24, 48, and 72 h). RESULTS: There was a significant and sustained increase in plasma IL-6 levels at all time points when compared with baseline values. Paired values were used in the statistical analysis. Median plasma (interquartile range) values of IL-6 were elevated from 15.5 (9-52) pg/ml at baseline to 23 (4-48) pg/ml (P < 0.01) at 8 h; from 15 (2-48) pg/ml at baseline to 46 (21-105) pg/ml (P < 0.001) at 24 h; from 16 (9-52) pg/ml at baseline to 61 (32-170) pg/ml (P < 0.001) at 48 h; and from 15.5 (6-48) pg/ml to 68 (40-200) pg/ml (P < 0.001) at 72 h. Significance was maintained even when adjusted for multiple comparisons. The relative increase in IL-6 concentration was dose-dependent. Moreover, IL-6 elevation had a direct correlation with flavopiridol peak plasma concentration, flavopiridol area under the curve, and plasma C-Reactive protein levels. A significant decrease in plasma granulocyte macrophage colony-stimulating factor occurred at the 8-h sampling point: 50 pg/ml (interquartile range 10-205 pg/ml, P < 0.01) when compared with baseline plasma levels and 71 pg/ml (interquartile range 5-152 pg/ml, P < 0.01). No changes in the other pro or anti-inflammatory cytokines were observed. Immunohistochemistry studies in bone marrow aspirates from a prospective group of patients in this trial demonstrated approximately 4-fold induction of IL-6 (compared with baseline), mostly in non-T cells. CONCLUSION: Biochemical analysis of plasma in patients undergoing infusional flavopiridol found a significant dose-dependent induction of IL-6. IL-6 elevation could be a marker for the process leading to the appearance of the proinflammatory syndrome observed in patients treated with infusional flavopiridol. The mechanism(s) underlying IL-6 induction and its significance are still unknown but may influence strategies to modulate flavopiridol's clinical effects.


Asunto(s)
Antineoplásicos/efectos adversos , Flavonoides/efectos adversos , Inflamación/inducido químicamente , Interleucina-6/sangre , Neoplasias/tratamiento farmacológico , Piperidinas/efectos adversos , Antineoplásicos/administración & dosificación , Médula Ósea/patología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/efectos adversos , Flavonoides/administración & dosificación , Humanos , Infusiones Intravenosas , Interleucinas/sangre , Neoplasias/sangre , Neoplasias/inmunología , Piperidinas/administración & dosificación
19.
Adv Exp Med Biol ; 566: 59-65, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16594135

RESUMEN

Since conventional therapies are directly dependent on the supply of either drugs or oxygen, a key question is whether antiangiogenic agents produce detrimental effects on tumor vascular function, thus compromising combination therapies. A second question is whether experimental results based on fast-growing, transplanted tumors mimic those in slowly developing spontaneous tumors, which may be more representative of response in human primary tumors. To investigate changes in tumor pathophysiology, three antiangiogenic agents were compared: a) endostatin, b) anti-VEGFR-2 (DC101), and c) celecoxib. Total blood vessels were identified using anti-CD31, perfused vessels using DiOC7, and hypoxia by EF5 uptake. Although individual tumor growth rates varied substantially, DC101 produced the most striking inhibition. DC101 increased total and perfused vessel spacing as well as overall hypoxia, while endostatin increased total vessel spacing, and hypoxia and celecoxib had no marked effects. These results reinforce the idea that pathophysiological changes in spontaneous tumors are in general reflective of response in transplanted tumors. Furthermore, although DC101 inhibited growth in roughly half of the spontaneous tumors, the remaining tumors were unaffected. A key focus of future studies will be to investigate the underlying rationale for the widely varying antiangiogenic response among tumors that outwardly appear so similar.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Celecoxib , Endostatinas/farmacología , Femenino , Hipoxia/tratamiento farmacológico , Hipoxia/fisiopatología , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/fisiopatología , Ratones , Ratones Endogámicos C3H , Pirazoles/farmacología , Proteínas Recombinantes/farmacología , Sulfonamidas/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
20.
Adv Exp Med Biol ; 566: 249-56, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16594159

RESUMEN

Hyaluronan (HA), a large negatively-charged polysaccharide, is a major component of vessel basal membrane. HA is expressed by a variety of cells, including tumor and endothelial cells. We hypothesized that HA could be up-regulated by hypoxia to enhance vessel formation. To determine the effect of hypoxia on the production of HA, tumor cells were treated with either media alone (control) or a hypoxia inducer (CoCl or NaN3) for 24 h. The level of HA in the media was then measured by ELISA. The results showed that both CoCl and NaN3 induced the production of HA. Since the low molecular weight form of HA (SMW) possesses pro-angiogenic properties, we investigated whether hypoxia-induced HA can be processed into SMW. Under hypoxic conditions, the activity of hyaluronidase, the enzyme responsible for degrading HA, was measured by an ELISA-like assay. The activity of hyaluronidase was shown to be up-regulated by hypoxia and, further, could carry out the function of processing HA into SMW. In addition, the hypoxic areas of tumor tissues were stained strongly with biotinylated HA-binding proteins, indicating that the level of HA was high compared to the oxic areas. This study demonstrates that hypoxia can stimulate the production of HA and the activity of hyaluronidase, which may promote angiogenesis as a compensation mechanism for hypoxia.


Asunto(s)
Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Hipoxia/metabolismo , Neoplasias/metabolismo , Acetilglucosaminidasa/metabolismo , Animales , Antígenos de Neoplasias , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Histona Acetiltransferasas , Humanos , Ácido Hialurónico/biosíntesis , Ácido Hialurónico/química , Ratones , Ratones Endogámicos C3H , Peso Molecular , Proteínas de Neoplasias/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica , beta-N-Acetilhexosaminidasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA