Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Breast Cancer Res Treat ; 183(2): 333-346, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32683564

RESUMEN

PURPOSE: This proof-of-concept study investigates gene expression in core needle biopsies (CNB) to predict whether individuals diagnosed with ductal carcinoma in situ (DCIS) on CNB were affected by invasion at the time of diagnosis. METHODS: Using a QuantiGene Plex 2.0 assay, 14 gene expression profiling was performed in 303 breast tissue samples. Preoperative diagnostic performance of a gene was measured by area under receiver-operating characteristic curve (AUC) with 95% confidence interval (CI). The gene mRNA positivity cutoff was computed using Gaussian mixture model (GMM); protein expression was measured by immunohistochemistry; DNA methylation was evaluated by targeted bisulfite sequencing. RESULTS: mRNA from 69% (34/49) mammoplasties, 72% (75/104) CNB DCIS, and 89% (133/150) invasive breast cancers (IBC) were analyzed. Based on pre-and post-surgery DCIS chart reviews, 21 cases were categorized as DCIS synchronous with invasion and 54 DCIS were pure DCIS without pathologic evidence of invasive disease. The ectopic expression of neuronal cadherin CDH2 was probable in 0% mammoplasties, 6% pure DCIS, 29% synchronous DCIS, and 26% IBC. The CDH2 mRNA positivity in preoperative biopsies showing pure DCIS was predictive of a final diagnosis of invasion (AUC = 0.67; 95% CI 0.53-0.80; P = 0.029). Site-specific methylation of the CDH2 promoter (AUC = 0.76; 95% CI 0.54-0.97; P = 0.04) and measurements of N-cadherin, a pro-invasive cell-cell adhesion receptor encoded by CDH2 (AUC = 0.8; 95% CI 0.66-0.99; P < 0.005) had a discriminating power allowing for discernment of CDH2-positive biopsy. CONCLUSIONS: Evidence of CDH2/N-cadherin expression, predictive of invasion synchronous with DCIS, may help to clarify a diagnosis and direct the course of therapy earlier in a patient's care.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Detección Precoz del Cáncer/métodos , Regulación Neoplásica de la Expresión Génica , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/genética , Biomarcadores de Tumor/genética , Biopsia con Aguja Gruesa , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Cadherinas/genética , Carcinoma Intraductal no Infiltrante/diagnóstico , Carcinoma Intraductal no Infiltrante/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Persona de Mediana Edad , Invasividad Neoplásica , Curva ROC , Adulto Joven
2.
Biochem J ; 463(2): 257-70, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25028810

RESUMEN

The Ras-related GTPase Rap1 promotes cell adhesion and migration. Although the significance of Rap1 contribution to cell migration is increasingly being recognized, little is known about the biochemical mechanisms driving this process. In the present study, we discovered a previously unidentified regulatory role of insulin-like growth factor type I (IGF-I) receptor (IGF-IR) in CRK Src homology 3 (SH3)-binding guanine-nucleotide-releasing protein (C3G)-Rap1-fascin-actin axis promoting cell movement. We demonstrate that a burst of Rap1 activity, rather than presumed hyperactivation, is imperative for the onset of cell movement. We show that while autophosphorylated IGF-IR signals to C3G to activate Rap1, subsequent IGF-IR internalization promotes gradual inactivation of Rap1 by putative Rap1 GTPase-activating protein (GAP). Additionally, IGF-IR signalling recruits active Rap1 at sites of cell motile protrusions. C3G depletion prevents IGF-I-induced fascin accumulation at actin microspikes and blocks protrusions. In the absence of IGF-IR activity, the wild-type (WT) Rap1 and the constitutively active V12Rap1 mutant remain in cell-cell contacts. Forced inactivation of Rap1 signalling by overexpressing dominant negative N17Rap1, Rap1GAP or by silencing C3G has a detrimental effect on filamentous (F)-actin and cell adhesion irrespective of IGF-IR signalling. We conclude that the basal levels of Rap1 activity holds up cell adhesion, whereas sequential regulation of C3G and GAP by IGF-IR reverses the labile Rap1 function from supporting adhesion to promoting migration.


Asunto(s)
Adhesión Celular , Movimiento Celular , Células Epiteliales/enzimología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor 2 Liberador de Guanina Nucleótido/genética , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Células MCF-7 , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap1/genética
3.
Cancer Res ; 79(13): 3492-3502, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31113820

RESUMEN

In the era of omics-driven research, it remains a common dilemma to stratify individual patients based on the molecular characteristics of their tumors. To improve molecular stratification of patients with breast cancer, we developed the Gaussian mixture model (GMM)-based classifier. This probabilistic classifier was built on mRNA expression data from more than 300 clinical samples of breast cancer and healthy tissue and was validated on datasets of ESR1, PGR, and ERBB2, which encode standard clinical markers and therapeutic targets. To demonstrate how a GMM approach could be exploited for multiclass classification using data from a candidate marker, we analyzed the insulin-like growth factor I receptor (IGF1R), a promising target, but a marker of uncertain importance in breast cancer. The GMM defined subclasses with downregulated (40%), unchanged (39%), upregulated (19%), and overexpressed (2%) IGF1R levels; inter- and intrapatient analyses of IGF1R transcript and protein levels supported these predictions. Overexpressed IGF1R was observed in a small percentage of tumors. Samples with unchanged and upregulated IGF1R were differentiated tumors, and downregulation of IGF1R correlated with poorly differentiated, high-risk hormone receptor-negative and HER2-positive tumors. A similar correlation was found in the independent cohort of carcinoma in situ, suggesting that loss or low expression of IGF1R is a marker of aggressiveness in subsets of preinvasive and invasive breast cancer. These results demonstrate the importance of probabilistic modeling that delves deeper into molecular data and aims to improve diagnostic classification, prognostic assessment, and treatment selection. SIGNIFICANCE: A GMM classifier demonstrates potential use for clinical validation of markers and determination of target populations, particularly when availability of specimens for marker development is low.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/clasificación , Modelos Estadísticos , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Invasividad Neoplásica , Pronóstico , Receptor ErbB-2/genética , Receptor IGF Tipo 1/genética , Receptores de Estrógenos/genética , Receptores de Progesterona/genética
4.
Int J Cancer ; 123(2): 464-475, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18449880

RESUMEN

This study investigates the role of tumor nitric oxide (NO) and vascular regulation in tumor ulceration following high-dose tumor necrosis factor-alpha (TNF) treatment. Using TNF-responsive (MethA) and nonresponsive (LL2) mouse tumors, tumor NO concentration was measured with an electrochemical sensor and tumor blood flow by Doppler ultrasound. Mice were also pretreated with a selective inducible nitric oxide synthase (iNOS) inhibitor, 1400 W. Tumors harvested from TNF-treated mice were cryosectioned and immunostained for murine macrophages, or/and iNOS. MethA tumor-bearing mice were depleted of macrophages. Pre- and post-TNF tumor NO levels were measured continuously, and mice were followed for gross tumor response. In MethA tumors, TNF caused a 96% response rate, and tumor NO concentration doubled. Tumor blood flow decreased to 3% of baseline by 4 hr and was sustained at 24 hr and 10 days post-TNF. Selective NO inhibition with 1400 W blocked NO rise and decreased response rate to 38%. MethA tumors showed tumor infiltration by macrophages post-TNF and the pattern of macrophage immunostaining overlapped with iNOS immunostaining. Depletion of macrophages inhibited tumor NO increase and response to TNF. LL2 tumors had a 0% response rate to TNF and exhibited no change in NO concentration. Blood flow decreased to 2% of baseline by 4 hr, recovered to 56% by 24 hr and increased to 232% by 10 days. LL2 tumors showed no infiltration by macrophages post-TNF. We conclude that TNF causes tumor infiltrating, macrophage-derived iNOS-mediated tumor NO rise and sustained tumor blood flow shutdown, resulting in tumor ulceration in the responsive tumor.


Asunto(s)
Antineoplásicos/farmacología , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Ácido Clodrónico/administración & dosificación , Ácido Clodrónico/farmacología , Selectina E/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrosarcoma/enzimología , Fibrosarcoma/metabolismo , Regulación Enzimológica de la Expresión Génica , Inmunohistoquímica , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Flujo Sanguíneo Regional , Molécula 1 de Adhesión Celular Vascular/metabolismo
5.
Cancer Res ; 63(21): 7232-40, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14612518

RESUMEN

Tumor oxygen status is a reliable prognostic marker that impacts malignant progression and outcome of tumor therapy. However, tumor oxygenation is heterogeneous and cannot be sufficiently described by a single parameter. It is influenced by several factors including microvessel density (MVD), blood flow (BF), blood volume (BV), blood oxygen saturation, tissue pO(2), oxygen consumption rate, and hypoxic fraction. The goal of this investigation was to integrate these measurements to obtain a comprehensive profile of tumor oxygenation. Platelet/endothelial cell adhesion molecule immunohistochemistry, the recessed oxygen microelectrode, color and power Doppler ultrasound (DUS), and diffuse light spectroscopy (DLS) were used to measure tumor oxygen status using vascular endothelial growth factor (VEGF)-transfected hypervascular human melanoma xenografts and their nontransfected counterparts as a model. NIH1286 human melanoma cells were transfected with a retroviral vector +/- a 720-bp fragment of human VEGF(121). High VEGF-producing clones were selected by ELISA. Oxygen consumption rate was measured in NIH1286/VEGF+ [VEGF-transfected cells (VEGF+ cells)] and NIH1286/Vec cells [cells transfected with vector alone (Vec cells)] using a standard Clark oxygen electrode. Athymic nude 6-8-week-old mice received s.c. injection in the right flank with 5 x 10(6) VEGF+ or Vec cells. When tumors were 10-14 mm in maximum dimension, serum was analyzed for VEGF by ELISA. Cryopreserved tumor tissue sections were immunostained for platelet/endothelial cell adhesion molecule, and MVD measurements were made. Tumor-bearing mice were anesthetized, and pO(2) measurements were made using Eppendorf pO(2) histograph or the recessed oxygen microelectrode. Tumor BF and BV were measured by quantitative analysis of DUS images. DLS was used to measure tumor BF and blood oxygen saturation variation. VEGF+ cell supernatants had 15,500 pg/ml VEGF, and Vec cells had 10 pg/ml. VEGF+ and Vec cells had equivalent oxygen consumption rates. VEGF+ tumors had a faster growth rate than Vec tumors. Serum from VEGF+ tumor-bearing mice showed 4,211 pg/ml VEGF, whereas VEGF was undetectable in the serum of control mice. MVD values were 74 +/- 11 in VEGF+ tumors and 39 +/- 4 in control tumors at x200 magnification/0.95-mm(2) area. The median pO(2) values were 3.5-fold higher in VEGF+ tumors than in Vec tumors by the recessed oxygen microelectrode and 18-fold higher by Eppendorf pO(2) histograph. DUS showed a 3.3-fold higher mean BF and a 5.5-fold higher BV in VEGF+ tumors than in Vec tumors. DLS showed a 3.2-fold higher mean BF and 1.7-fold higher oxygen saturation in the hypervascular tumors as compared with the control tumor type, consistent with increased BF and BV data by DUS. An integrated approach that yields a comprehensive and consistent profile of oxygen status in tumors could potentially provide critical information for prognosis and treatment.


Asunto(s)
Melanoma/irrigación sanguínea , Oxígeno/sangre , Animales , Línea Celular Tumoral , Humanos , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Oxígeno/metabolismo , Presión Parcial , Transfección , Trasplante Heterólogo , Ultrasonografía Doppler/métodos , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
6.
J Clin Endocrinol Metab ; 90(6): 3440-8, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15797967

RESUMEN

CONTEXT: Currently, there is no effective treatment for iodine-resistant thyroid cancers. OBJECTIVE: As a new approach to treatment, the efficacy of replication-selective, human thyroglobulin (TG) enhancer and promoter-driven, adenovirus (AdhTGEP)-mediated oncolysis was investigated using two well-differentiated thyroid cancer cell lines, XTC (TG positive) and FTC-133 (TG negative), and other control tumor and nontumor cell lines (all TG negative). DESIGN: A cohort study design was used. SETTING: The study setting was laboratory bench-top experiments. SUBJECTS/PARTICIPANTS: In vitro TG-expressing and nonexpressing thyroid cell culture lines, nonthyroid tumor cell lines, as well as preclinical thyroid tumor-bearing mice were studied. INTERVENTION: Adenoviral infection of cell lines was determined by immunohistochemistry, selective replication by one-step growth assays, and cytotoxicity by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphophenyl)-2H-tetrozolium (MTS) assay. In vivo tumor growth inhibition was determined by a single intratumoral injection of 1 x 10(9) plaque-forming units AdhTGEP, AdLacZ (control virus), or PBS to 50- to 75-mm(3) tumors. XTC cells showed intense immunohistochemical staining, whereas FTC-133 and all other control cell lines showed minimal staining for viral infection with AdhTGEP. MAIN OUTCOME MEASURES: Cell survival and tumor growth inhibition after adenoviral infection were the main outcome measures. RESULTS: One-step growth assays showed at least a more than 60-fold titer of AdhTGEP in XTC than in FTC-133 cells. Cytotoxicity assays showed approximately 68% cell kill in XTC and minimal cell kill in FTC-133 and all other control cell lines at a multiplicity of infection of 250. There was significant in vivo growth inhibition of AdhTGEP-treated XTC tumors (67 +/- 49 mm(3)) compared with AdLacZ-treated XTC (228 +/- 45 mm(3); P < 0.01), PBS-treated XTC (372 +/- 70 mm(3); P < 0.001), or AdhTGEP-treated FTC-133 tumors (598 +/- 168 mm(3)). CONCLUSION: Replication-selective virus-mediated oncolysis is a potential therapy for recurrent, well-differentiated, TG-secreting thyroid cancer that is unresponsive to standard treatment.


Asunto(s)
Adenoviridae/fisiología , Elementos de Facilitación Genéticos , Terapia Genética , Regiones Promotoras Genéticas , Tiroglobulina/genética , Neoplasias de la Tiroides/terapia , Adenoviridae/genética , Secuencia de Bases , Línea Celular , Supervivencia Celular/efectos de los fármacos , Estudios de Cohortes , Cartilla de ADN , Humanos , Neoplasias de la Tiroides/patología , Transfección , Replicación Viral
7.
Surgery ; 135(4): 427-36, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15041967

RESUMEN

BACKGROUND: Interferon-beta (IFNbeta) has multiple antitumor effects; however, its use has been limited by its short half-life in vivo. This limitation may be overcome by IFNbeta gene therapy. We evaluated adenovirus-IFNbeta therapy in an immunocompetent mouse model of carcinomatosis. METHODS: Mice that were treated intraperitoneally 5 days after tumor (mouse ovarian teratoma) inoculation with an adenoviral vector that contains the mouse IFNbeta gene (Ad-IFNbeta), control adenoviral vector or saline solution. Mice were monitored for multiple outcome measures and toxicity. To determine the mechanism of antitumor effect, flow cytometry of ascites fluid was performed to differentiate immune cell populations. Nitric oxide in ascites fluid was measured with an electrochemical microsensor. RESULTS: Tumor burden was decreased and survival was prolonged (P<.001) in the Ad-IFNbeta group after a single treatment of 3.3 x 10(8) plaque-forming units, with acceptable toxicity. By flow cytometry, an increase in the proportion of natural killer cells (from less than 2% of the gated population to more than 8%; P=.024) and an increase in macrophages were seen in the treated animals. Although there was a trend toward increased levels of nitric oxide in Ad-IFNbeta treatment groups, it was not statistically significant. CONCLUSION: IFNbeta gene therapy results in decreased tumor burden and improved survival in an aggressive, immunocompetent mouse model of carcinomatosis. This therapy warrants further evaluation as a treatment for disseminated peritoneal cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Terapia Genética/métodos , Interferón beta/uso terapéutico , Neoplasias Ováricas/terapia , Teratoma/terapia , Adenoviridae , Animales , Línea Celular Tumoral , Femenino , Vectores Genéticos , Inmunocompetencia , Ratones , Ratones Endogámicos C3H , Modelos Animales , Neoplasias Ováricas/patología , Teratoma/patología
8.
Biomark Res ; 2: 22, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25785189

RESUMEN

BACKGROUND: A subset of patients with ductal carcinoma in situ (DCIS) will develop invasive breast cancer (IBC). To date, there are no effective predictive biomarkers for identifying this subset with worse prognosis whose lesions are essentially indistinguishable histologically from those with favorable outcomes. We hypothesized that measurable parameters that discriminate DCIS from DCIS with concurrent invasion may serve as diagnostic biomarkers (BM) of progressive cancer in situ (CIS). RESULTS: Using a novel imaging-based method of tissue testing, we measured the relative expression levels of three candidate BM proteins specifically implicated in IBC progression - the insulin-like growth factor I receptor (IGF-IR), Ras-related protein 1 (Rap1), and Vav2 oncoprotein. Protein profiles were compared in 42 histologically normal mammary epithelial samples, 71 CIS (35 without/36 with invasion either on diagnostic biopsy or final surgical excision), and 98 IBC of known estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) status. The levels of the IGF-IR and Rap1 protein expression were significantly elevated in ER-positive (ER+/PR+/-/HER2 -) DCIS relative to normal epithelium (P <0.0001). The IGF-IR protein expression was also significantly up regulated in HER2-positive (ER+/-/PR+/-/HER2+) DCIS relative to normal epithelium (P = 0.0002). IGF-IR and Rap1 protein expression levels were similar among DCIS patients without or with concurrent invasion. Vav2 upregulation in DCIS relative to normal group was not associated with steroid hormone receptor and HER2 status, but was associated with the presence of concurrent invasion, including microinvasion (invasive foci of less than 1 mm). DCIS with high Vav2 were more than twice as likely to progress to invasive cancers as DCIS with low Vav2 (odds ratio, 2.42; 95% CI, 1.26-4-65; P =0.008). Furthermore, a receiver operating characteristic curve analysis revealed moderate ability of Vav2 protein expression measurements in DCIS to predict the existence of invasion concurrent with DCIS (area under the curve, 0.71; 95% CI, 0.59- 0.84). CONCLUSIONS: Our novel findings hold promise for utilizing Vav2 protein as a predictive BM for differentiating progressive from non-progressive DCIS.

9.
J Thyroid Res ; 2011: 979840, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22046576

RESUMEN

The development of molecular biomarkers (BMs) of follicular thyroid carcinoma is aimed at advancing diagnosis of follicular neoplasm, as histological examination of those tumors does not lend itself to definitive diagnosis of carcinoma. We assessed the relative levels of expression of 6 genes: CCND2, PCSK2, PLAB, RAP2A, TSHR, and IGF-1R in archived thyroid tissue. The quantitative real-time PCR analysis revealed a significant change in 3 genes: PSCK2 (a 22.4-fold decrease, P = 2.81E - 2), PLAB (an 8.3-fold increase, P = 9.81E - 12), and RAP2A (a 6.3-fold increase, P = 9.13E - 10) in carcinoma compared with adenoma. Expression of PCSK2 was equally low, PLAB was equally high, whereas RAP2A expression was significantly higher (25.9-fold, P = 0.039) in microdissected carcinoma cells that have invaded through the thyroid capsule and entered blood vessels than in thyroid tumor cells growing under the capsule. Thus, RAP2A appeared as a unique and worthy of further evaluation candidate BM associated with invasion of thyroid follicular cells.

10.
Ann Surg Oncol ; 9(4): 411-8, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11986195

RESUMEN

BACKGROUND: Mature dendritic cells (DCs) are potent antigen-presenting cells that activate naive T lymphocytes and initiate cellular immune responses. The ability of CD83(+) mature DCs infected with vaccinia virus encoding the gp100 melanoma transgene (rV-gp100) to stimulate an antimelanoma CD8(+) T-cell response was investigated. METHODS: Monocyte-derived immature or CD83(+) mature DCs were infected with rV-gp100. The activation state of the DCs and the expression of gp100 protein were evaluated by flow cytometry. The reactivity of antimelanoma CD8(+) T cells was confirmed by measuring specific interferon gamma secretion by using enzyme-linked immunosorbent assay in a mixed-tumor lymphocyte culture. RESULTS: Both immature and CD83(+) mature DCs expressed gp100 protein when the DCs were infected with rV-gp100. Calcium-signaling agents were required to induce maturation of both infected and noninfected immature DCs. Only rV-gp100-infected CD83(+) DCs induced CD8(+) T cells, after a single stimulation that recognized both peptide-pulsed target cells to multiple gp100 epitopes and a melanoma cell line that endogenously expressed gp100 antigen. CONCLUSIONS: CD83(+) DCs transduced with rV-gp100 are capable of generating a strong CD8(+) T-cell response against melanoma tumor cells. Expression of melanoma antigens by mature DCs offers the potential advantage of presenting multiple endogenously processed T-cell epitopes and using multiple HLA restriction elements for antimelanoma vaccine therapy.


Asunto(s)
Presentación de Antígeno/genética , Antígeno HLA-A2/genética , Inmunoglobulinas/genética , Melanoma/inmunología , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Virus Vaccinia/genética , Antígenos CD , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Citotoxicidad Inmunológica/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Antígeno HLA-A2/metabolismo , Humanos , Inmunoglobulinas/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de Neoplasias/inmunología , Células Tumorales Cultivadas , Virus Vaccinia/inmunología , Antígeno gp100 del Melanoma , Antígeno CD83
11.
Am J Physiol Heart Circ Physiol ; 284(1): H317-29, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12388254

RESUMEN

High-dose TNF with melphalan has significant antitumor activity in regional perfusion of the limbs and liver in human malignancies. TNF is believed to target tumor vasculature, but the precise molecular mechanism is unknown. The present study demonstrates that TNF downregulates the VEGF receptor, fetal liver kinase-1 (Flk-1), on tumor endothelium in a human melanoma xenograft model. NIH1286 human melanoma cells were transduced with a 720-bp fragment of the human VEGF(121) gene to develop well-vascularized tumors that served as an amplified system for measuring Flk-1 expression changes. We injected 5 x 10(6) cells subcutaneously, each of two distinct single cell clones (NIH1286/3 and NIH1286/15), into athymic nude mice to produce tumors approximately 10 mm in size. Each animal then received either BSA or TNF in BSA by tail vein. Tumors harvested at different time points post-TNF were analyzed for Flk-1 mRNA and protein expression. Data obtained showed that intravascular TNF downregulated Flk-1 expression in tumor endothelial cells. This effect could contribute to the antitumor activity of TNF known to target tumor vasculature.


Asunto(s)
Melanoma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Regulación hacia Abajo , Factores de Crecimiento Endotelial/genética , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Linfocinas/genética , Melanoma/genética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , ARN Mensajero/metabolismo , Albúmina Sérica Bovina/farmacología , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA