Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Metastasis Rev ; 33(1): 345-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24375391

RESUMEN

Neuroendocrine tumors (NETs) represent a heterogeneous group of diseases with varied natural history and prognosis depending upon the organ of origin and grade of aggressiveness. The most widely used biomarker to determine disease burden and monitor response to treatment is chromogranin A (CgA), but it is far from being the optimal predictive and prognostic biomarker in NETs. Biological understanding and derived treatment options for NETs have changed markedly in recent years. Over the last decade, the genomic landscape of these tumors has been extensively investigated. This has resulted in the discovery of mutations and expression anomalies in genes and pathways such as the PI3K/Akt/mTOR, DAXX/ATRX, and MEN1, which are promising predictive and prognostic biomarkers and future candidates for targeted therapies. Additionally, the study of tumor stroma and environment are one of the most promising fields for discovery of potential new targets and biomarkers.


Asunto(s)
Biomarcadores de Tumor/genética , Predisposición Genética a la Enfermedad/genética , Mutación , Tumores Neuroendocrinos/genética , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Humanos , Terapia Molecular Dirigida/métodos , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Neoplasma ; 59(6): 719-27, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22862173

RESUMEN

There is emerging evidence that cancer stem cells (CSCs), like normal tissue stem cells, are regulated by a niche formed of mesenchymal cells. In this review we summarize the current knowledge of the role of cancer associated fibroblasts (CAFs) in a tentative CSC niche. We also discuss findings from our own studies showing that CAF derived factors have a strong stimulatory effect on the stem cell properties of breast cancer cells. Based on recent literature we conclude that CAFs are strong modulators of the stem cell properties of cancer cells. This effect is likely to be particularly relevant under circumstances of early stages of tumor cell dissemination and metastasis.


Asunto(s)
Fibroblastos/fisiología , Neoplasias/patología , Células Madre Neoplásicas/fisiología , Animales , Humanos , Metástasis de la Neoplasia , Fenotipo
3.
Nat Cell Biol ; 3(5): 512-6, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11331881

RESUMEN

The term 'platelet-derived growth factor' (PDGF) refers to a family of disulphide-bonded dimeric isoforms that are important for growth, survival and function in several types of connective tissue cell. So far, three different PDGF chains have been identified - the classical PDGF-A and PDGF-B and the recently identified PDGF-C. PDGF isoforms (PDGF-AA, AB, BB and CC) exert their cellular effects by differential binding to two receptor tyrosine kinases. The PDGF alpha-receptor (PDGFR-alpha) binds to all three PDGF chains, whereas the beta-receptor (PDGFR-beta) binds only to PDGF-B. Gene-targeting studies using mice have shown that the genes for PDGF-A and PDGF-B, as well as the two PDGFR genes, are essential for normal development. Furthermore, overexpression of PDGFs is linked to different pathological conditions, including malignancies, atherosclerosis and fibroproliferative diseases. Here we have identify and characterize a fourth member of the PDGF family, PDGF-D. PDGF-D has a two-domain structure similar to PDGF-C and is secreted as a disulphide-linked homodimer, PDGF-DD. Upon limited proteolysis, PDGF-DD is activated and becomes a specific agonistic ligand for PDGFR-beta. PDGF-DD is the first known PDGFR-beta-specific ligand, and its unique receptor specificity indicates that it may be important for development and pathophysiology in several organs.


Asunto(s)
Linfocinas , Factor de Crecimiento Derivado de Plaquetas/química , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/química , Secuencia de Aminoácidos , Animales , Baculoviridae/metabolismo , Northern Blotting , Clonación Molecular , Cisteína/química , ADN Complementario/metabolismo , Dimerización , Relación Dosis-Respuesta a Droga , Humanos , Inmunohistoquímica , Insectos , Ligandos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Distribución Tisular
4.
Horm Metab Res ; 43(12): 884-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21960459

RESUMEN

The successful introduction of rationally targeted agents into standard cancer care is a testimony of the vast knowledge base in tumor biology. However, in order to provide individually tailored therapy to patients and to identify small subsets of patients with a high likelihood to benefit from treatment, the identification of biomarkers for response or resistance to a particular therapeutic regimen is imperative. Herein, by the use of a genetically engineered mouse model of pancreatic neuroendocrine tumors, we have assessed the utility of pericyte characteristics in terms of differential marker expression to serve as surrogate markers for response or evasive resistance to anti-angiogenic therapy. We found that tumors refractory to therapy following long-term treatment with a vascular endothelial growth factor receptor-2 blocking antibody contained blood vessels with a prolific investment of pericytes expressing α-smooth muscle actin. Further analysis by simultaneous immunostaining for different pericyte markers led to the conclusion that the increased abundance of this particular subtype of blood vessels most likely occurred by co-option of vessels from the surrounding exocrine pancreas. Our findings may form the basis for retrospective analysis of pancreatic neuroendocrine tumors from patients having undergone treatment with anti-angiogenic agents in order to validate the occurrence of pericytes expressing α-smooth muscle actin as a biomarker for tumors refractory to therapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Biomarcadores de Tumor/metabolismo , Resistencia a Antineoplásicos , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Pericitos/metabolismo , Pericitos/patología , Actinas/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Antígenos/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Tumores Neuroendocrinos/irrigación sanguínea , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pericitos/efectos de los fármacos , Proteoglicanos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Coloración y Etiquetado , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Cancer Res ; 61(7): 2929-34, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11306470

RESUMEN

Most solid malignancies display interstitial hypertension and a poor uptake of anticancer drugs. Platelet-derived growth factor (PDGF) and the cognate tyrosine kinase receptors are expressed in many tumors. Signaling through PDGFbeta receptors was shown recently to increase interstitial fluid pressure (IFP) in dermis after anaphylaxis-induced lowering of IFP. In this study, we show that treatment with the selective PDGF receptor kinase inhibitor, STI571, formerly known as CGP57148B, decreased the interstitial hypertension and increased capillary-to-interstitium transport of 51Cr-EDTA in s.c. growing rat PROb colonic carcinomas. Furthermore, treatment with an antagonistic PDGF-B oligonucleotide aptamer decreased interstitial hypertension in these tumors. PDGFbeta receptors were expressed in blood vessels and stromal cells but not in the tumor cells of PROb colonic carcinomas. Our study indicates a previously unrecognized role of PDGF receptors in tumor biology, although similar effects of PDGF on IFP have been demonstrated previously in the dermis. The data suggest interference with PDGF receptors, or their ligands, as a novel strategy to increase drug uptake and therapeutic effectiveness of cancer chemotherapy.


Asunto(s)
Neoplasias del Colon/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Benzamidas , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Neoplasias del Colon/irrigación sanguínea , Ácido Edético/farmacocinética , Inhibidores Enzimáticos/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Mesilato de Imatinib , Microdiálisis , Oligonucleótidos/farmacología , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , Ratas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Especificidad por Sustrato , Receptor 1 de Factores de Crecimiento Endotelial Vascular
6.
Cancer Res ; 61(15): 5778-83, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11479215

RESUMEN

Dermatofibrosarcoma protuberans (DFSP) and giant cell fibroblastoma (GCF) are recurrent, infiltrative skin tumors that presently are treated with surgery. DFSP and GCF tumors are genetically characterized by chromosomal rearrangements fusing the collagen type Ialpha1 (COLIA1) gene to the platelet-derived growth factor B-chain (PDGFB) gene. It has been shown that the resulting COL1A1/PDGF-B fusion protein is processed to mature PDGF-BB. Autocrine PDGF receptor stimulation has therefore been predicted to contribute to DFSP and GCF tumor development and growth. Here we demonstrate presence of activated PDGF receptors in primary cultures derived from six different DFSP and GCF tumors. Three of the primary cultures were further characterized; their in vitro growth displayed an increased sensitivity to treatment with the PDGF receptor tyrosine kinase inhibitor STI571, as compared with normal fibroblasts. Transplantable tumors, displaying a DFSP-like histology, were established from one of the DFSP primary cultures. Treatment of tumor-bearing severe combined immunodeficient mice with STI571 reduced tumor growth. The growth-inhibitory effects in vitro and in vivo occurred predominantly through induction of tumor cell apoptosis. Our study demonstrates growth-inhibitory effects of PDGF receptor antagonists on human DFSP- and GCF-derived tumor cells and demonstrates that autocrine PDGF receptor stimulation provides antiapoptotic signals contributing to the growth of these cells. These findings suggest targeting of PDGF receptors as a novel treatment strategy for DFSP and GCF.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Dermatofibrosarcoma/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Adulto , Animales , Benzamidas , División Celular/efectos de los fármacos , División Celular/fisiología , Preescolar , Dermatofibrosarcoma/irrigación sanguínea , Dermatofibrosarcoma/tratamiento farmacológico , Femenino , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Tumores de Células Gigantes/irrigación sanguínea , Tumores de Células Gigantes/tratamiento farmacológico , Tumores de Células Gigantes/patología , Inhibidores de Crecimiento/farmacología , Humanos , Mesilato de Imatinib , Lactante , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 59(15): 3719-23, 1999 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10446987

RESUMEN

Dermatofibrosarcoma protuberans (DFSP) displays chromosomal rearrangements involving chromosome 17 and 22, which fuse the collagen type Ialpha1 (COLIA1) gene to the platelet-derived growth factor (PDGF) B-chain (PDGFB) gene. To characterize the functional and structural properties of the COLIA1/PDGFB fusion protein, we generated a stable NIH3T3 cell line that contained a tumor-derived chimeric gene resulting from a COIA1 intron 7-PDGFB intron 1 fusion. Expression of the fusion protein led to morphological transformation and increased growth rate of these cells. The PDGF receptor kinase inhibitor CGP57148B reversed the transformed phenotype and reduced the growth rate of COLIA1/PDGFB-expressing cells but had no effects on control cells. The presence of dimeric COLIA1/PDGFB precursors was demonstrated through PDGFB immunoprecipitations of metabolically labeled cells and also by PDGFB immunoprecipitations followed by immunoblotting with COLIA1 antibodies. Pulse-chase studies demonstrated that the COLIA1/PDGFB precursor was processed to an end product that was indistinguishable from wild-type PDGF-BB. Finally, COLIA1/PDGFB-expressing cells generated tumors after s.c. injection into nude mice, and tumor growth was reduced by treatment with CGP57148B. We conclude that the COLIA1/PDGFB fusion associated with DFSP contributes to tumor development through ectopic production of PDGF-BB and the formation of an autocrine loop. Our findings, thus, suggest that PDGF receptors could be a target for pharmacological treatment of DFSP and giant cell fibroblastoma, e.g., through the use of PDGF receptor kinase inhibitors such as CGP57148B.


Asunto(s)
Dermatofibrosarcoma/genética , Proteínas de Fusión Oncogénica/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Neoplasias Cutáneas/genética , Células 3T3 , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Becaplermina , Benzamidas , Transformación Celular Neoplásica/genética , Cósmidos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Mesilato de Imatinib , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Proteínas de Fusión Oncogénica/genética , Piperazinas/farmacología , Piperazinas/uso terapéutico , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Proteínas Proto-Oncogénicas c-sis , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología
8.
Oncogene ; 35(39): 5119-31, 2016 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-26999641

RESUMEN

Therapy directed against oncogenic FLT3 has been shown to induce response in patients with acute myeloid leukemia (AML), but these responses are almost always transient. To address the mechanism of FLT3 inhibitor resistance, we generated two resistant AML cell lines by sustained treatment with the FLT3 inhibitor sorafenib. Parental cell lines carry the FLT3-ITD (tandem duplication) mutation and are highly responsive to FLT3 inhibitors, whereas resistant cell lines display resistance to multiple FLT3 inhibitors. Sanger sequencing and protein mass-spectrometry did not identify any acquired mutations in FLT3 in the resistant cells. Moreover, sorafenib treatment effectively blocked FLT3 activation in resistant cells, whereas it was unable to block colony formation or cell survival, suggesting that the resistant cells are no longer FLT3 dependent. Gene expression analysis of sensitive and resistant cell lines, as well as of blasts from patients with sorafenib-resistant AML, suggested an enrichment of the PI3K/mTOR pathway in the resistant phenotype, which was further supported by next-generation sequencing and phospho-specific-antibody array analysis. Furthermore, a selective PI3K/mTOR inhibitor, gedatolisib, efficiently blocked proliferation, colony and tumor formation, and induced apoptosis in resistant cell lines. Gedatolisib significantly extended survival of mice in a sorafenib-resistant AML patient-derived xenograft model. Taken together, our data suggest that aberrant activation of the PI3K/mTOR pathway in FLT3-ITD-dependent AML results in resistance to drugs targeting FLT3.


Asunto(s)
Resistencia a Antineoplásicos/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Morfolinas/administración & dosificación , Niacinamida/análogos & derivados , Compuestos de Fenilurea/administración & dosificación , Triazinas/administración & dosificación , Tirosina Quinasa 3 Similar a fms/genética , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Mutación , Niacinamida/administración & dosificación , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Sorafenib , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Oncogene ; 35(43): 5585-5596, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27065333

RESUMEN

Cartilage oligomeric matrix protein (COMP) is a soluble pentameric protein expressed in cartilage and involved in collagen organization. Tissue microarrays derived from two cohorts of patients with breast cancer (n=122 and n=498) were immunostained, revealing varying expression of COMP, both in the tumor cells and surrounding stroma. High levels of COMP in tumor cells correlated, independently of other variables, with poor survival and decreased recurrence-free survival. Breast cancer cells, MDA-MB-231, stably expressing COMP were injected into the mammary fat pad of SCID (CB-17/Icr-Prkdcscid/Rj) mice. Tumors expressing COMP were significantly larger and were more prone to metastasize as compared with control, mock-transfected, tumors. In vitro experiments confirmed that COMP-expressing cells had a more invasive phenotype, which could in part be attributed to an upregulation of matrix metalloprotease-9. Furthermore, microarray analyses of gene expression in tumors formed in vivo showed that COMP expression induced higher expression of genes protecting against endoplasmic reticulum stress. This observation was confirmed in vitro as COMP-expressing cells showed better survival as well as a higher rate of protein synthesis when treated with brefeldin A, compared with control cells. Further, COMP-expressing cells appeared to undergo a metabolic switch, that is, a Warburg effect. Thus, in vitro measurement of cell respiration indicated decreased mitochondrial metabolism. In conclusion, COMP is a novel biomarker in breast cancer, which contributes to the severity of the disease by metabolic switching and increasing invasiveness and tumor cell viability, leading to reduced survival in animal models and human patients.


Asunto(s)
Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Transformación Celular Neoplásica/metabolismo , Animales , Apoptosis/genética , Biomarcadores de Tumor , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Proteína de la Matriz Oligomérica del Cartílago/genética , Adhesión Celular/genética , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular/genética , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Inmunohistoquímica , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones SCID , Metástasis de la Neoplasia , Fosforilación Oxidativa , Pronóstico , Modelos de Riesgos Proporcionales , Recurrencia
10.
Arterioscler Thromb Vasc Biol ; 20(11): E89-95, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11073860

RESUMEN

Several antagonists specific for platelet-derived growth factor (PDGF) or its receptors have recently been developed and shown to inhibit intimal hyperplasia formation in various animal models, but data investigating the durability of this intervention is limited. The present study was designed to investigate the potency of PDGF B-chain aptamer, a novel type of PDGF-AB and -BB antagonist, in the rat carotid model and to characterize intermediate-term effects on lesion formation. One hundred thirty-four animals were randomized to aptamer treatment or placebo. Daily treatment with the antagonist resulted in a 50% reduction in lesion size at 2 weeks (P<0.001). The beneficial effect involved increased apoptosis and possibly an interference with smooth muscle cell migration. Discontinuing administration 1 week earlier did not give any significant benefit compared with phosphate-buffered saline-treated controls. When the antagonist was administered for 2 weeks and the vessels analyzed 6 weeks later, the beneficial effect was lost and the treated lesions had a higher intima-media and area-cell ratio compared with the treated lesions in the 2-week-endpoint study. Our findings confirm a role of PDGF B-chain in intimal hyperplasia, but the successful use of PDGF antagonists may require either prolonged treatment or combination therapy with other agents.


Asunto(s)
Arterias Carótidas/patología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Túnica Íntima/patología , Angioplastia de Balón/efectos adversos , Animales , Becaplermina , Sitios de Unión/efectos de los fármacos , Arterias Carótidas/química , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Hiperplasia , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Oligonucleótidos/metabolismo , Oligonucleótidos/farmacología , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Recurrencia , Túnica Íntima/química , Túnica Íntima/efectos de los fármacos , Túnica Íntima/metabolismo
11.
Oncogene ; 32(50): 5614-24, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23955085

RESUMEN

During breast cancer progression, transforming growth factor-beta (TGF-ß) switches from acting as a growth inhibitor to become a major promoter of epithelial-mesenchymal transition (EMT), invasion and metastasis. However, the mechanisms involved in this switch are not clear. We found that loss of CCAAT-enhancer binding protein beta (C/EBPß), a differentiation factor for the mammary epithelium, was associated with signs of EMT in triple-negative human breast cancer, and in invasive areas of mammary tumors in MMTV-PyMT mice. Using an established model of TGF-ß-induced EMT in mouse mammary gland epithelial cells, we discovered that C/EBPß was repressed during EMT by miR-155, an oncomiR in breast cancer. Depletion of C/EBPß potentiated the TGF-ß response towards EMT, and contributed to evasion of the growth inhibitory response to TGF-ß. Furthermore, loss of C/EBPß enhanced invasion and metastatic dissemination of the mouse mammary tumor cells to the lungs after subcutaneous injection into mice. The mechanism by which loss of C/EBPß promoted the TGF-ß response towards EMT, invasion and metastasis, was traced to a previously uncharacterized role of C/EBPß as a transcriptional activator of genes encoding the epithelial junction proteins E-cadherin and coxsackie virus and adenovirus receptor. The results identify miR-155-mediated loss of C/EBPß as a mechanism, which promotes breast cancer progression by shifting the TGF-ß response from growth inhibition to EMT, invasion and metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteína beta Potenciadora de Unión a CCAAT/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , Factor de Crecimiento Transformador beta/farmacología , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , MicroARNs/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Invasividad Neoplásica , Metástasis de la Neoplasia , Unión Proteica , Interferencia de ARN , Activación Transcripcional
12.
J Mol Biol ; 407(2): 298-315, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21277312

RESUMEN

Platelet-derived growth factor receptor (PDGFR) ß is a marker of stromal pericytes and fibroblasts and represents an interesting target for both diagnosis and therapy of solid tumors. A receptor-specific imaging agent would be a useful tool for further understanding the prognostic role of this receptor in vivo. Affibody molecules constitute a class of very small binding proteins that are highly suited for in vivo imaging applications and that can be selected to specifically recognize a desired target protein. Here we describe the isolation of PDGFRß-specific Affibody molecules with subnanomolar affinity. First-generation Affibody molecules were generated from a large naive library using phage display selection. Subsequently, sequences from binders having a desired selectivity profile and competing with the natural ligand for binding were used in the design of an affinity maturation library, which was created using a single partially randomized oligonucleotide. From this second-generation library, Affibody molecules with a 10-fold improvement in affinity (K(d)=0.4-0.5 nM) for human PDGFRß and a 4-fold improvement in affinity (K(d)=6-7 nM) for murine PDGFRß were isolated and characterized. Complete reversible folding after heating to 90 °C, as demonstrated by circular dichroism analysis, supports tolerance to labeling conditions for molecular imaging. The binders were highly specific, as verified by dot blot showing staining reactivity only with human and murine PDGFRß, but not with human PDGFRα, or a panel of control proteins including 16 abundant human serum proteins. The final binder recognized the native conformation of PDGFRß expressed in murine NIH-3T3 fibroblasts and human AU565 cells, and inhibited ligand-induced receptor phosphorylation in PDGFRß-transfected porcine aortic endothelial cells. The PDGFRß-specific Affibody molecule also accumulated around tumoral blood vessels in a model of spontaneous insulinoma, confirming a potential for in vivo targeting.


Asunto(s)
Anticuerpos/metabolismo , Ingeniería de Proteínas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología , Animales , Afinidad de Anticuerpos/inmunología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Femenino , Humanos , Ratones , Células 3T3 NIH , Neoplasias/diagnóstico , Biblioteca de Péptidos , Unión Proteica , Porcinos
13.
Oncogene ; 29(30): 4276-86, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20498640

RESUMEN

The Notch ligand delta-like ligand 4 (DLL4) is an essential component expressed by endothelial tip cells during angiogenic sprouting. We have described a conceptually novel therapeutic strategy for targeting tumor angiogenesis and endothelial tip cells based on DNA vaccination against DLL4. Immunization with DLL4-encoding plasmid DNA by in vivo electroporation severely retarded the growth of orthotopically implanted mammary carcinomas in mice by induction of a nonproductive angiogenic response. Mechanistically, vaccination brought about a break in tolerance against the self-antigen, DLL4, as evidenced by the production of inhibitory and inherently therapeutic antibodies against mouse DLL4. Importantly, no evidence for a delayed wound healing response, or for toxicity associated with pharmacological blockade of DLL4 signaling, was noted in mice immunized with the DLL4 vaccine. We have thus developed a well-tolerated DNA vaccination strategy targeting the endothelial tip cells and the antigen DLL4 with proven therapeutic efficacy in mouse models of mammary carcinoma; a disease that has been reported to dramatically induce the expression of DLL4. Conceivably, induction of immunity toward principal mediators of pathological angiogenesis could provide protection against recurrent malignant disease in the adjuvant setting.


Asunto(s)
Neoplasias Mamarias Experimentales/terapia , Proteínas de la Membrana/inmunología , Neovascularización Patológica/prevención & control , Vacunas de ADN/inmunología , Animales , Electroquimioterapia , Femenino , Inmunización , Interferón gamma/biosíntesis , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Cicatrización de Heridas
14.
Oncogene ; 28(4): 534-44, 2009 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18997817

RESUMEN

Members of the platelet-derived growth factor (PDGF) family are mitogens for cells of mesenchymal origin and have important functions during embryonic development, blood vessel maturation, fibrotic diseases and cancer. In contrast to the two classical PDGFs, the novel and less well-characterized members, PDGF-CC and PDGF-DD, are latent factors that need to be processed extracellularly by activating proteases, before they can mediate PDGF receptor activation. Here, we elucidate the structural requirements for urokinase plasminogen activator (uPA)-mediated activation of PDGF-DD, as well as the intricate interplay with uPA receptor (uPAR) signalling. Furthermore, we show that activated PDGF-DD, in comparison to latent, more potently transforms NIH/3T3 cells in vitro. Conversely, xenograft studies in nude mice demonstrate that cells expressing latent PDGF-DD are more tumorigenic than those expressing activated PDGF-DD. These findings imply that a fine-tuned proteolytic activation, in the local milieu, controls PDGF-DD bioavailability. Moreover, we suggest that proteolytic activation of PDGF-DD reveals a retention motif mediating interactions with pericellular components. Our proposed mechanism, where uPA not only generates active PDGF-DD, but also regulates its spatial distribution, provides novel insights into the biological function of PDGF-DD.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Linfocinas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Secuencias de Aminoácidos/genética , Animales , Disponibilidad Biológica , Células COS , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Chlorocebus aethiops , Femenino , Humanos , Linfocinas/genética , Linfocinas/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Trasplante Heterólogo , Activador de Plasminógeno de Tipo Uroquinasa/genética
15.
J Cell Biochem ; 100(1): 1-15, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16888779

RESUMEN

The Mexican axolotl, Ambystoma mexicanum, is an excellent animal model for studying heart development because it carries a naturally occurring recessive genetic mutation, designated gene c, for cardiac nonfunction. The double recessive mutants (c/c) fail to form organized myofibrils in the cardiac myoblasts resulting in hearts that fail to beat. Tropomyosin expression patterns have been studied in detail and show dramatically decreased expression in the hearts of homozygous mutant embryos. Because of the direct interaction between tropomyosin and troponin T (TnT), and the crucial functions of TnT in the regulation of striated muscle contraction, we have expanded our studies on this animal model to characterize the expression of the TnT gene in cardiac muscle throughout normal axolotl development as well as in mutant axolotls. In addition, we have succeeded in cloning the full-length cardiac troponin T (cTnT) cDNA from axolotl hearts. Confocal microscopy has shown a substantial, but reduced, expression of TnT protein in the mutant hearts when compared to normal during embryonic development.


Asunto(s)
Ambystoma mexicanum/metabolismo , Miocardio/metabolismo , Troponina T/metabolismo , Ambystoma mexicanum/embriología , Ambystoma mexicanum/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Embrión no Mamífero/metabolismo , Inmunoquímica , Datos de Secuencia Molecular , Contracción Muscular , Mutación , Miocardio/citología , Unión Proteica , Homología de Secuencia de Aminoácido , Tropomiosina/metabolismo , Troponina T/genética
16.
Neurol Sci ; 24(2): 70-3, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12827542

RESUMEN

Platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (FGF-2) are involved in the development of grade 2 gliomas. The aim of the present study was to determine the presence of these growth factors in the cerebrospinal fluid (CSF) and to assess their usefulness as biological markers. CSF was collected from 7 adult patients with newly diagnosed supratentorial low-grade gliomas by lumbar puncture and was analysed together with matched serum samples using radioreceptor and enzyme-linked immunosorbant assays. Neither PDGF nor VEGF were detected in the CSF, and FGF-2 was measurable at extremely low concentrations in only 2 of 7 patients. Serum levels were within normal limits. We conclude that these growth factors are not released into the CSF in any significant amounts and are therefore not suitable as biological markers in grade 2 gliomas.


Asunto(s)
Factores de Crecimiento Endotelial/análisis , Glioma/líquido cefalorraquídeo , Sustancias de Crecimiento/líquido cefalorraquídeo , Péptidos y Proteínas de Señalización Intercelular/análisis , Linfocinas/análisis , Factor de Crecimiento Derivado de Plaquetas/análisis , Adolescente , Adulto , Análisis Químico de la Sangre/métodos , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/líquido cefalorraquídeo , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Glioma/sangre , Sustancias de Crecimiento/sangre , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
17.
J Cell Sci ; 108 ( Pt 1): 333-42, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7537746

RESUMEN

Angiotensin II is a potent vasoconstrictor that has been also implicated in vascular hyperproliferative diseases, including atherosclerosis and restenosis following angioplasty. Treatment of cultured, serum-starved rat aortic smooth muscle cells with angiotensin II causes rapid protein tyrosine phosphorylation that precedes cell mitogenesis. We have identified two of the phosphoproteins as paxillin (75 kilodaltons) and the tyrosine kinase pp125Fak, both components of actin-associated focal adhesion sites. Angiotensin II stimulated a 5-fold increase in the tyrosine phosphorylation of paxillin and a smaller (1.5-fold) increase in pp125Fak tyrosine phosphorylation. Paxillin tyrosine phosphorylation was evident within 1 minute, and was maximal after 10 minutes. Similar elevated protein tyrosine phosphorylation levels of paxillin were obtained with exposure of the rat aortic smooth muscle cells to peptides endothelin-1 and alpha-thrombin that function, as angiotensin II, through binding to members of the seven transmembrane domain G protein coupled receptors. Angiotensin II treatment also stimulated the production of a well-ordered actin-containing stress fiber network and prominent paxillin-containing focal adhesions. The focal adhesions stained intensely with anti-phosphotyrosine antibody suggesting the tyrosine phosphorylation of paxillin and cytoskeletal reorganization were tightly coupled. Angiotensin II receptor occupancy has been shown previously to lead to protein kinase C activation. However, compared to angiotensin II stimulation, a smaller, delayed increase in paxillin tyrosine phosphorylation was observed following direct protein kinase C activation by the phorbol ester phorbol 12-myristate-13-acetate. Paxillin tyrosine phosphorylation was selective for certain agonists since no increase in tyrosine phosphorylation of this protein was observed following exposure to the potent mitogen PDGF. Thus, actin-based cytoskeletal changes involving sites of cell adhesion to the extracellular matrix may play an important role in normal and pathophysiologic smooth muscle cell growth regulation in response to certain angiotensin II-type vasoactive agonists.


Asunto(s)
Angiotensina II/farmacología , Moléculas de Adhesión Celular/metabolismo , Adhesión Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Músculo Liso Vascular/fisiología , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/fisiología , Becaplermina , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Proteínas del Citoesqueleto/aislamiento & purificación , Endotelinas/farmacología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Cinética , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Paxillin , Fosfoproteínas/aislamiento & purificación , Fosforilación , Fosfotirosina , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Ratas , Receptor de Insulina/metabolismo , Proteínas Recombinantes/farmacología , Estrés Mecánico , Acetato de Tetradecanoilforbol/farmacología , Trombina/farmacología , Tirosina/análogos & derivados , Tirosina/análisis
18.
Biochemistry ; 39(9): 2370-5, 2000 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-10694405

RESUMEN

Platelet-derived growth factor (PDGF) is a dimeric growth factor acting through tyrosine kinase alpha- and beta-receptors. In both receptors, the extracellular parts are composed of five Ig-like domains. Functional mapping of the extracellular part of the receptors have shown that ligand-binding occurs to Ig-like domains 2 and 3 and that Ig-like domain 4 is involved in receptor-receptor interactions. Recombinant GST-fusion proteins of PDGF alpha-receptor Ig-like domains 1-4 and beta-receptor Ig-like domains 1-3 (alphaRD1-4-GST and betaRD1-3-GST) were generated and compared with their cleaved counterparts (alphaRD1-4 and betaRD1-3) with regard to their ability to block PDGF binding to cell surface receptors. In the case of both the alpha- and the beta-receptors, 100-1000-fold lower concentrations of the GST-fusion proteins were required, as compared to the cleaved forms, for inhibition of PDGF binding to cell surface receptors. alphaRD1-4-GST and betaRD1-3-GST, in contrast to alphaRD1-4 and betaRD1-3, were shown to occur as ligand independent dimers. Covalently cross-linked alphaRD1-4 dimers displayed a 50-fold increased potency as compared to alphaRD1-4. We thus conclude that the dimeric nature of alphaRD1-4-GST and betaRD1-3-GST is responsible for the high antagonistic potency of the fusion proteins.


Asunto(s)
Espacio Extracelular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Reactivos de Enlaces Cruzados/química , Dimerización , Espacio Extracelular/química , Espacio Extracelular/genética , Vectores Genéticos , Glutatión Transferasa/genética , Humanos , Ligandos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/química , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Solubilidad , Porcinos , Timidina/metabolismo , Transfección , Tritio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA