Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Mol Med ; 13(12): e12924, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34762341

RESUMEN

Long-range communication between tumor cells and the lymphatic vasculature defines competency for metastasis in different cancer types, particularly in melanoma. Nevertheless, the discovery of selective blockers of lymphovascular niches has been compromised by the paucity of experimental systems for whole-body analyses of tumor progression. Here, we exploit immunocompetent and immunodeficient mouse models for live imaging of Vegfr3-driven neolymphangiogenesis, as a versatile platform for drug screening in vivo. Spatiotemporal analyses of autochthonous melanomas and patient-derived xenografts identified double-stranded RNA mimics (dsRNA nanoplexes) as potent inhibitors of neolymphangiogenesis, metastasis, and post-surgical disease relapse. Mechanistically, dsRNA nanoplexes were found to exert a rapid dual action in tumor cells and in their associated lymphatic vasculature, involving the transcriptional repression of the lymphatic drivers Midkine and Vegfr3, respectively. This suppressive function was mediated by a cell-autonomous type I interferon signaling and was not shared by FDA-approved antimelanoma treatments. These results reveal an alternative strategy for targeting the tumor cell-lymphatic crosstalk and underscore the power of Vegfr3-lymphoreporters for pharmacological testing in otherwise aggressive cancers.


Asunto(s)
Melanoma , ARN Bicatenario , Animales , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Desnudos , Transducción de Señal
2.
Adv Drug Deliv Rev ; 175: 113833, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34147531

RESUMEN

Imaging of the lymphatic vasculature has gained great attention in various fields, not only because lymphatic vessels act as a key draining system in the body, but also for their implication in autoimmune diseases, organ transplant, inflammation and cancer. Thus, neolymphangiogenesis, or the generation of new lymphatics, is typically an early event in the development of multiple tumor types, particularly in aggressive ones such as malignant melanoma. Still, the understanding of how lymphatic endothelial cells get activated at distal (pre)metastatic niches and their impact on therapy is still unclear. Addressing these questions is of particular interest in the case of immune modulators, because endothelial cells may favor or halt inflammatory processes depending on the cellular context. Therefore, there is great interest in visualizing the lymphatic vasculature in vivo. Here, we review imaging tools and mouse models used to analyze the lymphatic vasculature during tumor progression. We also discuss therapeutic approaches based on nanomedicines to target the lymphatic system and the potential use of extracellular vesicles to track and target sentinel lymph nodes. Finally, we summarize main pre-clinical models developed to visualize the lymphatic vasculature in vivo, discussing their applications with a particular focus in metastatic melanoma.


Asunto(s)
Vesículas Extracelulares/metabolismo , Sistema Linfático/diagnóstico por imagen , Sistema de Administración de Fármacos con Nanopartículas , Animales , Vesículas Extracelulares/patología , Humanos , Ganglios Linfáticos/diagnóstico por imagen , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Sistema Linfático/efectos de los fármacos , Sistema Linfático/patología , Vasos Linfáticos/diagnóstico por imagen , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/patología , Ganglio Linfático Centinela/diagnóstico por imagen , Ganglio Linfático Centinela/patología
3.
Nat Med ; 26(12): 1865-1877, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33077955

RESUMEN

An open question in aggressive cancers such as melanoma is how malignant cells can shift the immune system to pro-tumorigenic functions. Here we identify midkine (MDK) as a melanoma-secreted driver of an inflamed, but immune evasive, microenvironment that defines poor patient prognosis and resistance to immune checkpoint blockade. Mechanistically, MDK was found to control the transcriptome of melanoma cells, allowing for coordinated activation of nuclear factor-κB and downregulation of interferon-associated pathways. The resulting MDK-modulated secretome educated macrophages towards tolerant phenotypes that promoted CD8+ T cell dysfunction. In contrast, genetic targeting of MDK sensitized melanoma cells to anti-PD-1/anti-PD-L1 treatment. Emphasizing the translational relevance of these findings, the expression profile of MDK-depleted tumors was enriched in key indicators of a good response to immune checkpoint blockers in independent patient cohorts. Together, these data reveal that MDK acts as an internal modulator of autocrine and paracrine signals that maintain immune suppression in aggressive melanomas.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Melanoma Experimental/terapia , Midkina/genética , Microambiente Tumoral/genética , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Terapia Genética , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Midkina/farmacología , FN-kappa B/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transcriptoma/genética
5.
Cancer Cell ; 35(1): 46-63.e10, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30581152

RESUMEN

Modulators of mRNA stability are not well understood in melanoma, an aggressive tumor with complex changes in the transcriptome. Here we report the ability of p62/SQSTM1 to extend mRNA half-life of a spectrum of pro-metastatic factors. These include FERMT2 and other transcripts with no previous links to melanoma. Transcriptomic, proteomic, and interactomic analyses, combined with validation in clinical biopsies and mouse models, identified a selected set of RNA-binding proteins (RBPs) recruited by p62, with IGF2BP1 as a key partner. This p62-RBP interaction distinguishes melanoma from other tumors where p62 controls autophagy or oxidative stress. The relevance of these data is emphasized by follow-up analyses of patient prognosis revealing p62 and FERMT2 as adverse determinants of disease-free survival.


Asunto(s)
Melanoma/metabolismo , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , ARN Mensajero/química , Proteínas de Unión al ARN/metabolismo , Proteína Sequestosoma-1/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Proteínas de la Membrana/química , Ratones , Proteínas de Neoplasias/química , Trasplante de Neoplasias , Mapas de Interacción de Proteínas , Proteómica/métodos , Estabilidad del ARN , Análisis de Matrices Tisulares
6.
Nat Commun ; 8(1): 2249, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29269732

RESUMEN

Melanomas are well-known for their altered mRNA expression profiles. Yet, the specific contribution of mRNA binding proteins (mRBPs) to melanoma development remains unclear. Here we identify a cluster of melanoma-enriched genes under the control of CUGBP Elav-like family member 1 (CELF1). CELF1 was discovered with a distinct prognostic value in melanoma after mining the genomic landscape of the 692 known mRBPs across different cancer types. Genome-wide transcriptomic, proteomic, and RNA-immunoprecipitation studies, together with loss-of-function analyses in cell lines, and histopathological evaluation in clinical biopsies, revealed an intricate repertoire of CELF1-RNA interactors with minimal overlap with other malignancies. This systems approach uncovered the oncogene DEK as an unexpected target and downstream effector of CELF1. Importantly, CELF1 and DEK were found to represent early-induced melanoma genes and adverse indicators of overall patient survival. These results underscore novel roles of CELF1 in melanoma, illustrating tumor type-restricted functions of RBPs in cancer.


Asunto(s)
Proteínas CELF1/fisiología , Melanoma/genética , Oncogenes , Biología de Sistemas , Regiones no Traducidas 3' , Biopsia , Proteínas CELF1/genética , Proteínas CELF1/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas Cromosómicas no Histona/metabolismo , Humanos , Inmunoprecipitación , Melanoma/patología , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Pronóstico , Proteómica , ARN Neoplásico/genética , Análisis de Supervivencia , Transcriptoma
7.
Nature ; 546(7660): 676-680, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28658220

RESUMEN

Cutaneous melanoma is a type of cancer with an inherent potential for lymph node colonization, which is generally preceded by neolymphangiogenesis. However, sentinel lymph node removal does not necessarily extend the overall survival of patients with melanoma. Moreover, lymphatic vessels collapse and become dysfunctional as melanomas progress. Therefore, it is unclear whether (and how) lymphangiogenesis contributes to visceral metastasis. Soluble and vesicle-associated proteins secreted by tumours and/or their stroma have been proposed to condition pre-metastatic sites in patients with melanoma. Still, the identities and prognostic value of lymphangiogenic mediators remain unclear. Moreover, our understanding of lymphangiogenesis (in melanomas and other tumour types) is limited by the paucity of mouse models for live imaging of distal pre-metastatic niches. Injectable lymphatic tracers have been developed, but their limited diffusion precludes whole-body imaging at visceral sites. Vascular endothelial growth factor receptor 3 (VEGFR3) is an attractive 'lymphoreporter' because its expression is strongly downregulated in normal adult lymphatic endothelial cells, but is activated in pathological situations such as inflammation and cancer. Here, we exploit this inducibility of VEGFR3 to engineer mouse melanoma models for whole-body imaging of metastasis generated by human cells, clinical biopsies or endogenously deregulated oncogenic pathways. This strategy revealed early induction of distal pre-metastatic niches uncoupled from lymphangiogenesis at primary lesions. Analyses of the melanoma secretome and validation in clinical specimens showed that the heparin-binding factor midkine is a systemic inducer of neo-lymphangiogenesis that defines patient prognosis. This role of midkine was linked to a paracrine activation of the mTOR pathway in lymphatic endothelial cells. These data support the use of VEGFR3 reporter mice as a 'MetAlert' discovery platform for drivers and inhibitors of metastasis.


Asunto(s)
Citocinas/metabolismo , Vasos Linfáticos/metabolismo , Metástasis de la Neoplasia/diagnóstico por imagen , Metástasis de la Neoplasia/patología , Imagen de Cuerpo Entero/métodos , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Femenino , Genes Reporteros , Humanos , Linfangiogénesis , Vasos Linfáticos/patología , Masculino , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Melanoma/patología , Ratones , Midkina , Comunicación Paracrina , Pronóstico , Recurrencia , Reproducibilidad de los Resultados , Serina-Treonina Quinasas TOR/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Cell ; 30(5): 694-707, 2016 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-27908735

RESUMEN

RNA binding proteins (RBPs) modulate cancer progression through poorly understood mechanisms. Here we show that the RBP UNR/CSDE1 is overexpressed in melanoma tumors and promotes invasion and metastasis. iCLIP sequencing, RNA sequencing, and ribosome profiling combined with in silico studies unveiled sets of pro-metastatic factors coordinately regulated by UNR as part of RNA regulons. In addition to RNA steady-state levels, UNR was found to control many of its targets at the level of translation elongation/termination. Key pro-oncogenic targets of UNR included VIM and RAC1, as validated by loss- and gain-of-function studies. Our results identify UNR as an oncogenic modulator of melanoma progression, unravel the underlying molecular mechanisms, and identify potential targets for this therapeutically challenging malignancy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Melanoma/patología , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba , Vimentina/genética , Proteína de Unión al GTP rac1/genética , Animales , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/genética , Ribosomas/genética , Análisis de Secuencia de ARN/métodos
9.
Oncotarget ; 7(48): 78971-78984, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27806339

RESUMEN

Vascular Endotelial Growth Factors C and D (VEGF-C and VEGF-D) are crucial regulators of lymphangiogenesis, a main event in the metastatic spread of breast cancer tumors. Although inhibition of lymphangiogenic gene expression might be a useful therapeutic strategy to restrict the progression of cancer, the factors involved in the transcriptional repression of these genes are still unknown. We have previously shown that Nuclear Receptor Corepressor 1 (NCoR) and the thyroid hormone receptor ß1 (TRß) inhibit tumor invasion. Here we show that these molecules repress VEGF-C and VEGF-D gene transcription in breast cancer cells, reducing lymphatic vessel density and sentinel lymph node invasion in tumor xenografts. The clinical significance of these results is stressed by the finding that NCoR and TRß transcripts correlate negatively with those of the lymphangiogenic genes and the lymphatic vessel marker LYVE-1 in human breast tumors. Our results point to the use of NCoR and TRß as potential biomarkers for diagnosis or prognosis in breast cancer and suggest that further studies of these molecules as potential targets for anti-lymphangiogenic therapy are warranted.


Asunto(s)
Neoplasias de la Mama/genética , Metástasis Linfática/patología , Co-Represor 1 de Receptor Nuclear/metabolismo , Receptores beta de Hormona Tiroidea/genética , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Co-Represor 1 de Receptor Nuclear/genética , Pronóstico , Transcripción Genética , Factor C de Crecimiento Endotelial Vascular/genética , Factor D de Crecimiento Endotelial Vascular/genética , Proteínas de Transporte Vesicular/genética
10.
Autophagy ; 12(10): 1776-1790, 2016 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-27464255

RESUMEN

Melanoma is a paradigm of aggressive tumors with a complex and heterogeneous genetic background. Still, melanoma cells frequently retain developmental traits that trace back to lineage specification programs. In particular, lysosome-associated vesicular trafficking is emerging as a melanoma-enriched lineage dependency. However, the contribution of other lysosomal functions such as autophagy to melanoma progression is unclear, particularly in the context of metastasis and resistance to targeted therapy. Here we mined a broad spectrum of cancers for a meta-analysis of mRNA expression, copy number variation and prognostic value of 13 core autophagy genes. This strategy identified heterozygous loss of ATG5 at chromosome band 6q21 as a distinctive feature of advanced melanomas. Importantly, partial ATG5 loss predicted poor overall patient survival in a manner not shared by other autophagy factors and not recapitulated in other tumor types. This prognostic relevance of ATG5 copy number was not evident for other 6q21 neighboring genes. Melanocyte-specific mouse models confirmed that heterozygous (but not homozygous) deletion of Atg5 enhanced melanoma metastasis and compromised the response to targeted therapy (exemplified by dabrafenib, a BRAF inhibitor in clinical use). Collectively, our results support ATG5 as a therapeutically relevant dose-dependent rheostat of melanoma progression. Moreover, these data have important translational implications in drug design, as partial blockade of autophagy genes may worsen (instead of counteracting) the malignant behavior of metastatic melanomas.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/genética , Pérdida de Heterocigocidad/genética , Melanoma/genética , Melanoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Mapeo Cromosómico , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Heterocigoto , Ratones , Metástasis de la Neoplasia , Nevo/genética , Nevo/patología , Pigmentación/genética , Pronóstico , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas B-raf/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Riesgo , Análisis de Supervivencia
12.
Angiogenesis ; 19(3): 433-45, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26993803

RESUMEN

The lymphatic system is essential in many physiological and pathological processes. Still, much remains to be known about the molecular mechanisms that control its development and function and how to modulate them therapeutically. The study of these mechanisms will benefit from better controlled genetic mouse models targeting specifically lymphatic endothelial cells. Among the genes expressed predominantly in lymphatic endothelium, Vegfr3 was the first one identified and is still considered to be one of the best lymphatic markers and a key regulator of the lymphatic system. Here, we report the generation of a Vegfr3-CreER (T2) knockin mouse by gene targeting in embryonic stem cells. This mouse expresses the tamoxifen-inducible CreER(T2) recombinase under the endogenous transcriptional control of the Vegfr3 gene without altering its physiological expression or regulation. The Vegfr3-CreER (T2) allele drives efficient recombination of floxed sequences upon tamoxifen administration specifically in Vegfr3-expressing cells, both in vitro, in primary lymphatic endothelial cells, and in vivo, at different stages of mouse embryonic development and postnatal life. Thus, our Vegfr3-CreER (T2) mouse constitutes a new powerful genetic tool for lineage tracing analysis and for conditional gene manipulation in the lymphatic endothelium that will contribute to improve our current understanding of this system.


Asunto(s)
Sistema Linfático/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen/métodos , Integrasas/genética , Sistema Linfático/citología , Sistema Linfático/crecimiento & desarrollo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Tamoxifeno/farmacología
13.
FEBS J ; 283(1): 25-38, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26443003

RESUMEN

A common feature of solid tumors is their ability to incite the formation of new blood and lymph vessels trough the processes of angiogenesis and lymphangiogenesis, respectively, to support tumor growth and favor metastatic dissemination. As a result of the lack of feedback regulatory control mechanisms or due to the exacerbated presence of pro-angiogenic signals within the tumor microenvironment, the tumor endothelium receives continuous signals to sprout and develop, generating vessels that are structurally and functionally abnormal. An emerging mechanism playing a central role in shaping the tumor vasculature is the endothelial-vesicular network that regulates trafficking/export and degradation of key signaling proteins and membrane receptors, including the vascular endothelial growth-factor receptor-2/3 and members of the Notch pathway. Here we will discuss recent evidence highlighting how vesicular trafficking mechanisms in endothelial cells contribute to pathological angiogenesis/lymphangiogenesis and can provide novel and exploitable targets in antiangiogenic therapies.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Animales , Células Endoteliales/patología , Humanos , Linfangiogénesis/efectos de los fármacos , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Transporte de Proteínas/efectos de los fármacos
14.
Int J Cancer ; 136(4): E62-73, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25178837

RESUMEN

Cell plasticity is emerging as a key regulator of tumor progression and metastasis. During carcinoma dissemination epithelial cells undergo epithelial to mesenchymal transition (EMT) processes characterized by the acquisition of migratory/invasive properties, while the reverse, mesenchymal to epithelial transition (MET) process, is also essential for metastasis outgrowth. Different transcription factors, called EMT-TFs, including Snail, bHLH and Zeb families are drivers of the EMT branch of epithelial plasticity, and can be post-transcriptionally downregulated by several miRNAs, as the miR-200 family. The specific or redundant role of different EMT-TFs and their functional interrelations are not fully understood. To study the interplay between different EMT-TFs, comprehensive gain and loss-of-function studies of Snail1, Snail2 and/or Zeb1 factors were performed in the prototypical MDCK cell model system. We here describe that Snail1 and Zeb1 are mutually required for EMT induction while continuous Snail1 and Snail2 expression, but not Zeb1, is needed for maintenance of the mesenchymal phenotype in MDCK cells. In this model system, EMT is coordinated by Snail1 and Zeb1 through transcriptional and epigenetic downregulation of the miR-200 family. Interestingly, Snail1 is involved in epigenetic CpG DNA methylation of the miR-200 loci, essential to maintain the mesenchymal phenotype. The present results thus define a novel functional interplay between Snail and Zeb EMT-TFs in miR-200 family regulation providing a molecular link to their previous involvement in the generation of EMT process in vivo.


Asunto(s)
Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/fisiología , MicroARNs/genética , Factores de Transcripción/fisiología , Animales , Metilación de ADN , Perros , Epigénesis Genética , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , MicroARNs/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
15.
Cancer Cell ; 26(1): 61-76, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24981740

RESUMEN

Although common cancer hallmarks are well established, lineage-restricted oncogenes remain less understood. Here, we report an inherent dependency of melanoma cells on the small GTPase RAB7, identified within a lysosomal gene cluster that distinguishes this malignancy from over 35 tumor types. Analyses in human cells, clinical specimens, and mouse models demonstrated that RAB7 is an early-induced melanoma driver whose levels can be tuned to favor tumor invasion, ultimately defining metastatic risk. Importantly, RAB7 levels and function were independent of MITF, the best-characterized melanocyte lineage-specific transcription factor. Instead, we describe the neuroectodermal master modulator SOX10 and the oncogene MYC as RAB7 regulators. These results reveal a unique wiring of the lysosomal pathway that melanomas exploit to foster tumor progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Linaje de la Célula , Lisosomas/enzimología , Melanoma/enzimología , Neoplasias Cutáneas/enzimología , Proteínas de Unión al GTP rab/metabolismo , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/mortalidad , Melanoma/secundario , Melanoma/terapia , Ratones , Invasividad Neoplásica , Estadificación de Neoplasias , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Factores de Tiempo , Transfección , Proteínas de Unión al GTP rab/genética , Proteínas de Unión a GTP rab7
16.
Proc Natl Acad Sci U S A ; 109(16): 6223-8, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22474390

RESUMEN

Lymphatic vessel growth or lymphangiogenesis occurs during embryonic development and wound healing and plays an important role in tumor metastasis and inflammatory diseases. However, the possibility of noninvasive detection and quantification of lymphangiogenesis has been lacking. Here, we present the Vegfr3(EGFPLuc) mouse model, where an EGFP-luciferase fusion protein, expressed under the endogenous transcriptional control of the Vegfr3 gene, allows the monitoring of physiological and pathological lymphangiogenesis in vivo. We show tracking of lymphatic vessel development during embryogenesis as well as lymphangiogenesis induced by specific growth factors, during wound healing and in contact hypersensitivity (CHS)--induced inflammation where we also monitor down-regulation of lymphangiogenesis by the glucocorticoid dexamethasone. Importantly, the Vegfr3-reporter allowed us to tracking tumor-induced lymphangiogenesis at the tumor periphery and in lymph nodes in association with the metastatic process. This is the first reporter mouse model for luminescence imaging of lymphangiogenesis. It should provide an important tool for studying the involvement of lymphangiogenesis in pathological processes.


Asunto(s)
Diagnóstico por Imagen/métodos , Inflamación/metabolismo , Vasos Linfáticos/metabolismo , Cicatrización de Heridas , Animales , Línea Celular Tumoral , Dexametasona/farmacología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Glucocorticoides/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inflamación/genética , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Linfangiogénesis/efectos de los fármacos , Metástasis Linfática , Vasos Linfáticos/embriología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factores de Tiempo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
Blood ; 119(19): 4565-76, 2012 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22446484

RESUMEN

Membrane-anchored ephrinB2 and its receptor EphB4 are involved in the formation of blood and lymphatic vessels in normal and pathologic conditions. Eph/ephrin activation requires cell-cell interactions and leads to bidirectional signaling pathways in both ligand- and receptor-expressing cells. To investigate the functional consequences of blocking ephrinB2 activity, 2 highly specific human single-chain Fv (scFv) Ab fragments against ephrinB2 were generated and characterized. Both Ab fragments suppressed endothelial cell migration and tube formation in vitro in response to VEGF and provoked abnormal cell motility and actin cytoskeleton alterations in isolated endothelial cells. As only one of them (B11) competed for binding of ephrinB2 to EphB4, these data suggest an EphB-receptor-independent blocking mechanism. Anti-ephrinB2 therapy reduced VEGF-induced neovascularization in a mouse Matrigel plug assay. Moreover, systemic administration of ephrinB2-blocking Abs caused a drastic reduction in the number of blood and lymphatic vessels in xenografted mice and a concomitant reduction in tumor growth. Our results show for the first time that specific Ab-based ephrinB2 targeting may represent an effective therapeutic strategy to be used as an alternative or in combination with existing antiangiogenic drugs for treating patients with cancer and other angiogenesis-related diseases.


Asunto(s)
Anticuerpos/farmacología , Efrina-B2/antagonistas & inhibidores , Linfangiogénesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Secuencia de Aminoácidos , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Anticuerpos/uso terapéutico , Especificidad de Anticuerpos , Regulación hacia Abajo/efectos de los fármacos , Efrina-B2/inmunología , Efrina-B2/metabolismo , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoterapia/métodos , Linfangiogénesis/fisiología , Ratones , Ratones Desnudos , Ratones SCID , Terapia Molecular Dirigida , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Genesis ; 49(1): 36-45, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21254335

RESUMEN

Cre/loxP-dependent expression of fluorescent proteins represents a powerful biological tool for cell lineage, fate-mapping, and genetic analysis. Live tissue imaging has significantly improved with the development of far-red fluorescent proteins, with optimized spectral characteristics for in vivo applications. Here, we report the generation of the first transgenic mouse line expressing the far-red fluorescent protein Katushka, driven by the hybrid CAG promoter upon Cre-mediated recombination. After germ line or tissue-specific Cre-driven reporter activation, Katushka expression is strong and ubiquitous, without toxic effects, allowing fluorescence detection in fresh and fixed samples from all tissues examined. Moreover, fluorescence can be detected by in vivo noninvasive whole-body imaging when Katuhska is expressed exclusively in a specific cell population deep within the animal body such as pancreatic beta cells. Thus, this reporter model enables early, widespread, and sensitive in vivo detection of Cre activity and should provide a versatile tool for a wide spectrum of fluorescence and live-imaging applications.


Asunto(s)
Integrasas/genética , Proteínas Luminiscentes/genética , Animales , Citometría de Flujo/métodos , Genes Reporteros , Ratones , Ratones Transgénicos , Microscopía Confocal/métodos , Regiones Promotoras Genéticas , Recombinación Genética , Proteína Fluorescente Roja
19.
Pharm Res ; 27(12): 2544-55, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20857179

RESUMEN

PURPOSE: To design hyaluronic acid (HA) and chitosan-g-poly(ethylene glycol) (CS-g-PEG) nanoparticles intended for a broad range of gene delivery applications. METHODS: Nanoparticles formulated at different HA/CS-g-PEG mass ratios were developed to associate either pDNA or siRNA. The physico-chemical characteristics, morphology, association efficiency and nuclease protection ability of the nanocarriers were compared for these two molecules. Their biological performance, including transfection effciency, nanoparticle cellular uptake and citotoxicity, was assesed. RESULTS: The resulting nanoparticles showed an adequate size (between 130 and 180 nm), and their surface charge could be modulated according to the nanoparticle composition (from +30 mV to -20 mV). All prototypes exhibited a greater association efficiency and nuclease protection for pDNA than for siRNA. However, cell culture experiments evidenced that HA/CS-g-PEG nanoparticles were effective carriers for the delivery of both, siRNA and pDNA, eliciting a biological response with minimal cytotoxicity. Moreover, experiments performed in the HEK-EGFP-Snail1 cell line showed the potential of the HA/CS-g-PEG nanoparticles to silence the expression of the Snail1 transcription factor, an important mediator in tumor progression. CONCLUSIONS: HA/CS-g-PEG nanoparticles can be easily modulated for the delivery of different types of gene molecules, offering great potential for gene therapy applications, as evidenced by their biological performance.


Asunto(s)
Quitosano/química , ADN/administración & dosificación , Terapia Genética , Ácido Hialurónico/química , Nanopartículas , Plásmidos , Polietilenglicoles/química , ARN Interferente Pequeño/administración & dosificación , Secuencia de Bases , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos
20.
Nat Protoc ; 4(11): 1591-613, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19834475

RESUMEN

Here we describe several methods for the characterization of epithelial-mesenchymal transition (EMT) at the cellular, molecular and behavioral level. This protocol describes both in vitro and in vivo approaches designed to analyze different features that when taken together permit the characterization of cells undergoing transient or stable EMT. We define straightforward methods for phenotypical, cellular and transcriptional characterization of EMT in vitro in monolayer cultures. The procedure also presents technical details for the generation of in vitro three-dimensional (3D) cultures analyzing cell phenotype and behavior during the EMT process. In addition, we describe xenotransplantation techniques to graft 3D cell cultures into mice to study in vivo invasion in a physiological-like environment. Finally, the protocol describes the analysis of selected EMT markers from experimental and human tumor samples. This series of methods can be applied to the study of EMT under various experimental and biological situations. Once the methodology is established, the time required to complete the protocol may vary from 3 to 4 weeks (monolayer cultures) and up to 6-8 weeks if including 3D cultures.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Epiteliales/citología , Mesodermo/citología , Animales , Secuencia de Bases , Desdiferenciación Celular , Diferenciación Celular , Cartilla de ADN/genética , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Humanos , Mesodermo/metabolismo , Mesodermo/trasplante , Ratones , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...