Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
NPJ Precis Oncol ; 6(1): 95, 2022 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-36575215

RESUMEN

Third-generation EGFR tyrosine kinase inhibitors (EGFR-TKIs), including osimertinib, an irreversible EGFR-TKI, are important treatments for non-small cell lung cancer with EGFR-TKI sensitizing or EGFR T790M resistance mutations. While patients treated with osimertinib show clinical benefit, disease progression and drug resistance are common. Emergence of de novo acquired resistance from a drug tolerant persister (DTP) cell population is one mechanism proposed to explain progression on osimertinib and other targeted cancer therapies. Here we profiled osimertinib DTPs using RNA-seq and ATAC-seq to characterize the features of these cells and performed drug screens to identify therapeutic vulnerabilities. We identified several vulnerabilities in osimertinib DTPs that were common across models, including sensitivity to MEK, AURKB, BRD4, and TEAD inhibition. We linked several of these vulnerabilities to gene regulatory changes, for example, TEAD vulnerability was consistent with evidence of Hippo pathway turning off in osimertinib DTPs. Last, we used genetic approaches using siRNA knockdown or CRISPR knockout to validate AURKB, BRD4, and TEAD as the direct targets responsible for the vulnerabilities observed in the drug screen.

2.
JCI Insight ; 6(19)2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34622806

RESUMEN

Myelodysplastic syndromes (MDS) are hematopoietic stem and progenitor cell (HSPC) malignancies characterized by ineffective hematopoiesis and an increased risk of leukemia transformation. Epigenetic regulators are recurrently mutated in MDS, directly implicating epigenetic dysregulation in MDS pathogenesis. Here, we identified a tumor suppressor role of the acetyltransferase p300 in clinically relevant MDS models driven by mutations in the epigenetic regulators TET2, ASXL1, and SRSF2. The loss of p300 enhanced the proliferation and self-renewal capacity of Tet2-deficient HSPCs, resulting in an increased HSPC pool and leukemogenicity in primary and transplantation mouse models. Mechanistically, the loss of p300 in Tet2-deficient HSPCs altered enhancer accessibility and the expression of genes associated with differentiation, proliferation, and leukemia development. Particularly, p300 loss led to an increased expression of Myb, and the depletion of Myb attenuated the proliferation of HSPCs and improved the survival of leukemia-bearing mice. Additionally, we show that chemical inhibition of p300 acetyltransferase activity phenocopied Ep300 deletion in Tet2-deficient HSPCs, whereas activation of p300 activity with a small molecule impaired the self-renewal and leukemogenicity of Tet2-deficient cells. This suggests a potential therapeutic application of p300 activators in the treatment of MDS with TET2 inactivating mutations.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Factores de Transcripción p300-CBP/genética , Animales , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epigénesis Genética , Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/metabolismo , Ratones , Mutación , Síndromes Mielodisplásicos/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Tasa de Supervivencia
3.
Sci Adv ; 6(23): eaaz7249, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32548262

RESUMEN

RING1B, a core Polycomb repressive complex 1 subunit, is a histone H2A ubiquitin ligase essential for development. RING1B is overexpressed in patients with luminal breast cancer (BC) and recruited to actively transcribed genes and enhancers co-occupied by the estrogen receptor α (ERα). Whether ERα-induced transcriptional programs are mediated by RING1B is not understood. We show that prolonged estrogen administration induces transcriptional output and chromatin landscape fluctuations. RING1B loss impairs full estrogen-mediated gene expression and chromatin accessibility for key BC transcription factors. These effects were mediated, in part, by RING1B enzymatic activity and nucleosome binding functions. RING1B is recruited in a cyclic manner to ERα, FOXA1, and GRHL2 cobound sites and regulates estrogen-induced enhancers and ERα recruitment. Last, ChIP exo revealed multiple binding events of these factors at single-nucleotide resolution, including RING1B occupancy approximately 10 base pairs around ERα bound sites. We propose RING1B as a key regulator of the dynamic, liganded-ERα transcriptional regulatory circuit in luminal BC.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Cromatina/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Complejo Represivo Polycomb 1/metabolismo
4.
Nucleic Acids Res ; 48(6): 2956-2968, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32025719

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) catalyzes the symmetric di-methylation of arginine residues in histones H3 and H4, marks that are generally associated with transcriptional repression. However, we found that PRMT5 inhibition or depletion led to more genes being downregulated than upregulated, indicating that PRMT5 can also act as a transcriptional activator. Indeed, the global level of histone H3K27me3 increases in PRMT5 deficient cells. Although PRMT5 does not directly affect PRC2 enzymatic activity, methylation of histone H3 by PRMT5 abrogates its subsequent methylation by PRC2. Treating AML cells with an EZH2 inhibitor partially restored the expression of approximately 50% of the genes that are initially downregulated by PRMT5 inhibition, suggesting that the increased H3K27me3 could directly or indirectly contribute to the transcription repression of these genes. Indeed, ChIP-sequencing analysis confirmed an increase in the H3K27me3 level at the promoter region of a quarter of these genes in PRMT5-inhibited cells. Interestingly, the anti-proliferative effect of PRMT5 inhibition was also partially rescued by treatment with an EZH2 inhibitor in several leukemia cell lines. Thus, PRMT5-mediated crosstalk between histone marks contributes to its functional effects.


Asunto(s)
Arginina/metabolismo , Histonas/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Transcripción Genética , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Eliminación de Gen , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Metilación , Ratones Noqueados , Modelos Biológicos , Nucleosomas/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores
5.
Cell Rep ; 24(10): 2643-2657, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30184499

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) is overexpressed in many cancer types and is a promising therapeutic target for several of them, including leukemia and lymphoma. However, we and others have reported that PRMT5 is essential for normal physiology. This dependence may become dose limiting in a therapeutic setting, warranting the search for combinatorial approaches. Here, we report that PRMT5 depletion or inhibition impairs homologous recombination (HR) DNA repair, leading to DNA-damage accumulation, p53 activation, cell-cycle arrest, and cell death. PRMT5 symmetrically dimethylates histone and non-histone substrates, including several components of the RNA splicing machinery. We find that PRMT5 depletion or inhibition induces aberrant splicing of the multifunctional histone-modifying and DNA-repair factor TIP60/KAT5, which selectively affects its lysine acetyltransferase activity and leads to impaired HR. As HR deficiency sensitizes cells to PARP inhibitors, we demonstrate here that PRMT5 and PARP inhibitors have synergistic effects on acute myeloid leukemia cells.


Asunto(s)
Proteína-Arginina N-Metiltransferasas/metabolismo , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Muerte Celular , Línea Celular Tumoral , Reparación del ADN/genética , Reparación del ADN/fisiología , Código de Histonas/genética , Código de Histonas/fisiología , Histonas/metabolismo , Humanos , Lisina Acetiltransferasa 5/genética , Lisina Acetiltransferasa 5/metabolismo , Lisina Acetiltransferasas/genética , Lisina Acetiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética
6.
Blood ; 129(20): 2782-2792, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28381396

RESUMEN

AML1-ETO (AE), a fusion oncoprotein generated by t(8;21), can trigger acute myeloid leukemia (AML) in collaboration with mutations including c-Kit, ASXL1/2, FLT3, N-RAS, and K-RAS. Caspase-3, a key executor among its family, plays multiple roles in cellular processes, including hematopoietic development and leukemia progression. Caspase-3 was revealed to directly cleave AE in vitro, suggesting that AE may accumulate in a Caspase-3-compromised background and thereby accelerate leukemogenesis. Therefore, we developed a Caspase-3 knockout genetic mouse model of AML and found that loss of Caspase-3 actually delayed AML1-ETO9a (AE9a)-driven leukemogenesis, indicating that Caspase-3 may play distinct roles in the initiation and/or progression of AML. We report here that loss of Caspase-3 triggers a conserved, adaptive mechanism, namely autophagy (or macroautophagy), which acts to limit AE9a-driven leukemia. Furthermore, we identify ULK1 as a novel substrate of Caspase-3 and show that upregulation of ULK1 drives autophagy initiation in leukemia cells and that inhibition of ULK1 can rescue the phenotype induced by Caspase-3 deletion in vitro and in vivo. Collectively, these data highlight Caspase-3 as an important regulator of autophagy in AML and demonstrate that the balance and selectivity between its substrates can dictate the pace of disease.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia , Carcinogénesis/patología , Caspasa 3/metabolismo , Leucemia/metabolismo , Leucemia/patología , Proteínas de Fusión Oncogénica/metabolismo , Animales , Antígenos CD34/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Autorrenovación de las Células , Modelos Animales de Enfermedad , Feto/patología , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Trasplante de Hígado , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , Especificidad por Sustrato
7.
AJR Am J Roentgenol ; 204(1): W11-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25539263

RESUMEN

OBJECTIVE: The purpose of this study was to evaluate the role of CT perfusion in monitoring response to neoadjuvant antiangiogenic and radiation therapy in resectable soft-tissue sarcomas and correlate the findings with tumor size, circulating and tumor biomarkers, and gene expression. SUBJECTS AND METHODS: This phase II clinical trial included 20 patients (13 men and 7 women; mean age, 55 years) with soft-tissue sarcomas who were undergoing treatment with the antiangiogenic drug bevacizumab followed by bevacizumab, radiation, and surgical resection. The patients underwent CT perfusion and diagnostic contrast-enhanced CT at baseline, at 2 weeks after bevacizumab therapy, and after completion of bevacizumab and radiation therapy. Multiple CT perfusion parameters (blood flow, blood volume, mean transit time, and permeability) were correlated with tumor size, circulating and tumor biomarkers, and gene expression. RESULTS: Two weeks after bevacizumab therapy, there was substantial fall in blood volume (31.9% reduction, p = 0.01) with more pronounced reduction in blood flow, blood volume, and permeability after treatment completion (53-64% reduction in blood flow, blood volume, and permeability; p = 0.001), whereas tumor size showed no significant change (p = 0.34). Tumors with higher baseline blood volume and lower baseline tumor size showed superior response to bevacizumab and radiation (p = 0.05). There was also an increase in median plasma vascular endothelial growth factor and placental-derived growth factor concentration after bevacizumab therapy paralleled by a decrease in tumor perfusion depicted by CT perfusion, although this was not statistically significant (p = 0.4). The baseline tumor microvessel density (MVD) correlated with blood flow (p = 0.04). At least 20 different genes were differentially expressed in tumors with higher and lower baseline perfusion. CONCLUSION: CT perfusion is more sensitive than tumor size for monitoring early and late response to bevacizumab and radiation therapy. CT perfusion parameters correlate with MVD, and the gene expression levels of baseline tumors could potentially predict treatment response.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Biomarcadores de Tumor/metabolismo , Quimioradioterapia/métodos , Imagen de Perfusión/métodos , Sarcoma/diagnóstico , Sarcoma/terapia , Tomografía Computarizada por Rayos X/métodos , Adulto , Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Bevacizumab , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante , Células Neoplásicas Circulantes/patología , Neovascularización Patológica/diagnóstico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Sarcoma/metabolismo , Resultado del Tratamiento
8.
Int J Cancer ; 132(1): 29-41, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22684860

RESUMEN

Increased levels of hypoxia and hypoxia-inducible factor 1α (HIF-1α) in human sarcomas correlate with tumor progression and radiation resistance. Prolonged antiangiogenic therapy of tumors not only delays tumor growth but may also increase hypoxia and HIF-1α activity. In our recent clinical trial, treatment with the vascular endothelial growth factor A (VEGF-A) antibody, bevacizumab, followed by a combination of bevacizumab and radiation led to near complete necrosis in nearly half of sarcomas. Gene Set Enrichment Analysis of microarrays from pretreatment biopsies found that the Gene Ontology category "Response to hypoxia" was upregulated in poor responders and that the hierarchical clustering based on 140 hypoxia-responsive genes reliably separated poor responders from good responders. The most commonly used chemotherapeutic drug for sarcomas, doxorubicin (Dox), was recently found to block HIF-1α binding to DNA at low metronomic doses. In four sarcoma cell lines, HIF-1α shRNA or Dox at low concentrations blocked HIF-1α induction of VEGF-A by 84-97% and carbonic anhydrase 9 by 83-93%. HT1080 sarcoma xenografts had increased hypoxia and/or HIF-1α activity with increasing tumor size and with anti-VEGF receptor antibody (DC101) treatment. Combining DC101 with HIF-1α shRNA or metronomic Dox had a synergistic effect in suppressing growth of HT1080 xenografts, at least in part via induction of tumor endothelial cell apoptosis. In conclusion, sarcomas respond to increased hypoxia by expressing HIF-1α target genes that may promote resistance to antiangiogenic and other therapies. HIF-1α inhibition blocks this evasive resistance and augments destruction of the tumor vasculature.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Sarcoma/terapia , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bevacizumab , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Hipoxia de la Célula/fisiología , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Doxorrubicina/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Terapia Genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Sarcoma/genética , Sarcoma/metabolismo , Sarcoma/patología , Factor A de Crecimiento Endotelial Vascular/genética
9.
Gastroenterology ; 141(1): 141-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21440548

RESUMEN

BACKGROUND & AIMS: A single nucleotide polymorphism 61*G (rs4444903) in the epidermal growth factor (EGF) gene has been associated, in 2 case-control studies, with hepatocellular carcinoma (HCC). We tested associations between demographic, clinical, and genetic data and development of HCC, and developed a simple predictive model in a cohort of patients with chronic hepatitis C and advanced fibrosis. METHODS: Black and white subjects from the Hepatitis C Antiviral Long-term Treatment against Cirrhosis (HALT-C) trial (n=816) were followed up prospectively for development of a definite or presumed case of HCC for a median time period of 6.1 years. We used the Cox proportional hazards regression model to determine the hazard ratio for risk of HCC and to develop prediction models. RESULTS: Subjects with EGF genotype G/G had a higher adjusted risk for HCC than those with genotype A/A (hazard ratio, 2.10; 95% confidence interval, 1.05-4.23; P=.03). After adjusting for EGF genotype, blacks had no increased risk of HCC risk compared with whites. Higher serum levels of EGF were observed among subjects with at least one G allele (P=.08); the subset of subjects with EGF G/G genotype and above-median serum levels of EGF had the highest risk of HCC. We developed a simple prediction model that included the EGF genotype to identify patients at low, intermediate, and high risk for HCC; 6-year cumulative HCC incidences were 2.3%, 10.4%, and 26%, respectively. CONCLUSIONS: We associated the EGF genotype G/G with increased risk for HCC; differences in its frequency among black and white subjects might account for differences in HCC incidence between these groups. We developed a model that incorporates EGF genotype and demographic and clinical variables to identify patients at low, intermediate, and high risk for HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Receptores ErbB/genética , Hepatitis C Crónica/genética , Cirrosis Hepática/genética , Neoplasias Hepáticas/genética , Polimorfismo de Nucleótido Simple , Adulto , Negro o Afroamericano/genética , Antivirales/uso terapéutico , Carcinoma Hepatocelular/etnología , Carcinoma Hepatocelular/virología , Progresión de la Enfermedad , Receptores ErbB/sangre , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Hepatitis C Crónica/complicaciones , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/etnología , Humanos , Incidencia , Estimación de Kaplan-Meier , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etnología , Cirrosis Hepática/virología , Neoplasias Hepáticas/etnología , Neoplasias Hepáticas/virología , Modelos Logísticos , Masculino , Persona de Mediana Edad , Fenotipo , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Estados Unidos , Población Blanca/genética
10.
Int J Radiat Oncol Biol Phys ; 81(4): 1081-90, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20932656

RESUMEN

PURPOSE: Numerous preclinical studies have demonstrated that angiogenesis inhibitors can increase the efficacy of radiotherapy (RT). We sought to examine the safety and efficacy of bevacizumab (BV) and RT in soft tissue sarcomas and explore biomarkers to help determine the treatment response. METHODS AND MATERIALS: Patients with ≥5 cm, intermediate- or high-grade soft tissue sarcomas at significant risk of local recurrence received neoadjuvant BV alone followed by BV plus RT before surgical resection. Correlative science studies included analysis of the serial blood and tumor samples and serial perfusion computed tomography scans. RESULTS: The 20 patients had a median tumor size of 8.25 cm, with 13 extremity, 1 trunk, and 6 retroperitoneal/pelvis tumors. The neoadjuvant treatment was well tolerated, with only 4 patients having Grade 3 toxicities (hypertension, liver function test elevation). BV plus RT resulted in ≥80% pathologic necrosis in 9 (45%) of 20 tumors, more than double the historical rate seen with RT alone. Three patients had a complete pathologic response. The median microvessel density decreased 53% after BV alone (p <.05). After combination therapy, the median tumor cell proliferation decreased by 73%, apoptosis increased 10.4-fold, and the blood flow, blood volume, and permeability surface area decreased by 62-72% (p <.05). Analysis of gene expression microarrays of untreated tumors identified a 24-gene signature for treatment response. The microvessel density and circulating progenitor cells at baseline and the reduction in microvessel density and plasma soluble c-KIT with BV therapy also correlated with a good pathologic response (p <.05). After a median follow-up of 20 months, only 1 patient had developed local recurrence. CONCLUSIONS: The results from the present exploratory study indicated that BV increases the efficacy of RT against soft tissue sarcomas and might reduce the incidence of local recurrence. Thus, this regimen warrants additional investigation. Gene expression profiles and other tissue and circulating biomarkers showed promising correlations with treatment response.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Sarcoma/terapia , Neoplasias de los Tejidos Blandos/terapia , Adulto , Anciano , Inhibidores de la Angiogénesis/efectos adversos , Anticuerpos Monoclonales Humanizados/efectos adversos , Bevacizumab , Biomarcadores de Tumor/sangre , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Microvasos/efectos de los fármacos , Microvasos/patología , Microvasos/efectos de la radiación , Persona de Mediana Edad , Terapia Neoadyuvante/efectos adversos , Terapia Neoadyuvante/métodos , Recurrencia Local de Neoplasia/prevención & control , Complicaciones Posoperatorias , Dosificación Radioterapéutica , Sarcoma/sangre , Sarcoma/irrigación sanguínea , Sarcoma/patología , Neoplasias de los Tejidos Blandos/irrigación sanguínea , Neoplasias de los Tejidos Blandos/patología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación
11.
Cancer Res ; 70(21): 8357-67, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20978198

RESUMEN

Tumors induce new blood vessel growth primarily from host organ microvascular endothelial cells (EC), and microvasculature differs significantly between the lung and liver. Vascular endothelial growth factor (VEGF or VEGF-A) promotion of tumor angiogenesis is thought to be mediated primarily by VEGF receptor-2 (VEGFR-2). In this study, VEGFR-2 antibody (DC101) inhibited growth of RenCa renal cell carcinoma lung metastases by 26%, whereas VEGFR-1 antibody (MF-1) had no effect. However, VEGFR-2 neutralization had no effect on RenCa liver metastases, whereas VEGFR-1 neutralization decreased RenCa liver metastases by 31%. For CT26 colon carcinoma liver metastases, inhibition of both VEGFR-1 and VEGFR-2 was required to induce growth delay. VEGFR-1 or VEGFR-2 inhibition decreased tumor burden not by preventing the establishment of micrometastases but rather by preventing vascularization and growth of micrometastases by 55% and 43%, respectively. VEGF induced greater phosphorylation of VEGFR-2 in lung ECs and of VEGFR-1 in liver ECs. EC proliferation, migration, and capillary tube formation in vitro were suppressed more by VEGFR-2 inhibition for lung EC and more by VEGFR-1 inhibition for liver EC. Collectively, our results indicate that liver metastases are more reliant on VEGFR-1 than lung metastases to mediate angiogenesis due to differential activity of VEGFRs on liver EC versus lung EC. Thus, therapies inhibiting specific VEGFRs should consider the targeted sites of metastatic disease.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Metástasis de la Neoplasia , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Western Blotting , Carcinoma de Células Renales/secundario , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Neoplasias del Colon/patología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Hemangiosarcoma/metabolismo , Hemangiosarcoma/patología , Hemangiosarcoma/prevención & control , Humanos , Técnicas para Inmunoenzimas , Neoplasias Renales/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/prevención & control , Especificidad de Órganos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...