Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jerónimo Blanco is active.

Publication


Featured researches published by Jerónimo Blanco.


Journal of Clinical Investigation | 2012

Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells

Toni Celià-Terrassa; Óscar Meca-Cortés; Francesca Mateo; Alexia Martínez de Paz; Nuria Rubio; Anna Arnal-Estapé; Brian Ell; Raquel Bermudo; Alba Díaz; Marta Guerra-Rebollo; Juan José Lozano; Conchi Estarás; Catalina Ulloa; Daniel ρlvarez-Simón; Jordi Milà; Ramón Vilella; Rosanna Paciucci; Marian A. Martínez-Balbás; Antonio García de Herreros; Roger R. Gomis; Yibin Kang; Jerónimo Blanco; Pedro L. Fernández; Timothy M. Thomson

Malignant progression in cancer requires populations of tumor-initiating cells (TICs) endowed with unlimited self renewal, survival under stress, and establishment of distant metastases. Additionally, the acquisition of invasive properties driven by epithelial-mesenchymal transition (EMT) is critical for the evolution of neoplastic cells into fully metastatic populations. Here, we characterize 2 human cellular models derived from prostate and bladder cancer cell lines to better understand the relationship between TIC and EMT programs in local invasiveness and distant metastasis. The model tumor subpopulations that expressed a strong epithelial gene program were enriched in highly metastatic TICs, while a second subpopulation with stable mesenchymal traits was impoverished in TICs. Constitutive overexpression of the transcription factor Snai1 in the epithelial/TIC-enriched populations engaged a mesenchymal gene program and suppressed their self renewal and metastatic phenotypes. Conversely, knockdown of EMT factors in the mesenchymal-like prostate cancer cell subpopulation caused a gain in epithelial features and properties of TICs. Both tumor cell subpopulations cooperated so that the nonmetastatic mesenchymal-like prostate cancer subpopulation enhanced the in vitro invasiveness of the metastatic epithelial subpopulation and, in vivo, promoted the escape of the latter from primary implantation sites and accelerated their metastatic colonization. Our models provide new insights into how dynamic interactions among epithelial, self-renewal, and mesenchymal gene programs determine the plasticity of epithelial TICs.


Laboratory Investigation | 2002

Combined noninvasive imaging and luminometric quantification of luciferase-labeled human prostate tumors and metastases.

Nadia El Hilali; Nuria Rubio; Mercedes Martinez-Villacampa; Jerónimo Blanco

Noninvasive imaging should facilitate the analysis of changes in experimental tumors and metastases-expressing photoproteins and result in improved data consistency and experimental animal welfare. We analyzed quantitative aspects of noninvasive imaging of luciferase-labeled tumors by comparing the efficiency of noninvasive light detection with in vitro quantification of luciferase activity. An intensified charge coupled device video camera was used to noninvasively image luciferase-expressing human prostate tumors and metastases in nude mice, after ip inoculation of luciferin. Repeated imaging of anesthetized animals after intervening growth periods allowed monitoring of tumor and metastases development. Comparison of photon events recorded in tumor images with the number of relative light units from luminometric quantification of homogenates from the same tumors, revealed that the efficiency with which light escapes tumors is inversely related to tumor size and that intensified charge coupled device images alone are not sufficient for quantitative evaluation of tumor growth. However, a combined videometric and luminometric approach did allow quantification and was used to show the cytostatic effects of paclitaxel in three different human prostate tumors growing in nude mice.


PLOS ONE | 2012

Human umbilical cord blood-derived mesenchymal stem cells promote vascular growth in vivo.

Santiago Roura; Juli R. Bagó; Carolina Soler-Botija; Josep Maria Pujal; Carolina Gálvez-Montón; Cristina Prat-Vidal; Aida Llucià-Valldeperas; Jerónimo Blanco; Antoni Bayes-Genis

Stem cell therapies are promising strategies to regenerate human injured tissues, including ischemic myocardium. Here, we examined the acquisition of properties associated with vascular growth by human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs), and whether they promoted vascular growth in vivo. UCBMSCs were induced in endothelial cell-specific growth medium (EGM-2) acquiring new cell markers, increased Ac-LDL uptake, and migratory capacity as assessed by qRT-PCR, Western blotting, indirect immunofluorescence, and invasion assays. Angiogenic and vasculogenic potentials could be anticipated by in vitro experiments showing self organization into Matrigel-mediated cell networks, and activation of circulating angiogenic-supportive myeloid cells. In mice, following subcutaneous co-injection with Matrigel, UCBMSCs modified to co-express bioluminescent (luciferases) and fluorescent proteins were demonstrated to participate in the formation of new microvasculature connected with the host circulatory system. Response of UCBMSCs to ischemia was explored in a mouse model of acute myocardial infarction (MI). UCBMSCs transplanted using a fibrin patch survived 4 weeks post-implantation and organized into CD31+network structures above the infarcted myocardium. MI-treated animals showed a reduced infarct scar and a larger vessel-occupied area in comparison with MI-control animals. Taken together, the presented results show that UCBMSCs can be induced in vitro to acquire angiogenic and vasculogenic properties and contribute to vascular growth in vivo.


Breast Cancer Research and Treatment | 2004

Overexpression of Bcl-xL in Human Breast Cancer Cells Enhances Organ-Selective Lymph Node Metastasis

Laura España; Yolanda Fernández; Nuria Rubio; Angels Torregrosa; Jerónimo Blanco; Angels Sierra

Lymph node metastasis are the first prognostic factor in breast cancer diagnosis and an early event in metastatic spread. To assess the role of anti-apoptotic proteins in lymph node metastatic progression of human breast cancer cells we analyzed the metastatic activity of MDA-MB-435 cells transfected with the Bcl-xL gene, after orthotopic inoculation in Nude Balb/c and in SCID mice. The luciferase gene was introduced by permanent transfection in the 435/Bcl-xL and 435/Neo cells and used as a tumor marker to measure the number of tumor cells lodged in lymph nodes. We found that 435/Bcl-xL tumor cells had enhanced organ-specific metastatic activity, preferentially lodging in peripheral lymph nodes, where at 45 days post-implantation we found 7 × 106± 6× 106 435/Bcl-xL.luc and 2 ±1.1 435/Neo.luc luciferase tagged tumor cell equivalents (TCEs). Metastases were abrogated in mice in which orthotopic tumors were induced with 435/Bcl-xL-antisense cells. Additionally, in vitro experiments show that in 435 cells Bcl-xL-antisense can override the emergence of resistance to apoptosis induced by TNF-α and TGF-β in cells overexpressing Bcl-xL, increasing also adhesion to extracellular matrix proteins. These results point to the relevance of Bcl-xL overexpression inducing lymph node metastasis of breast cancer cells, and to the value of this gene as a target for therapy in order to prevent metastasis.


Laboratory Investigation | 2001

Metastatic behavior of human breast carcinomas overexpressing the Bcl-x(L) gene: a role in dormancy and organospecificity.

Nuria Rubio; Laura España; Yolanda Fernández; Jerónimo Blanco; Angels Sierra

The ability of metastatic cells to survive antiapoptotic signals may contribute to the organospecific-spread patterns of clinical metastasis and dormancy. MDA-MB-435 breast cancer cells (435/Bcl-xL), which overexpress the Bcl-xL gene, were labeled with the luciferase gene and injected orthotopically into homozygous athymic Balb/c (nude) mice to study the metastatic behavior of the breast cancer cells. The overexpression of Bcl-xL in tumors increased the overall metastatic burden in mice (bones, liver, kidneys, brain, lungs, and lymph nodes) in comparison with control tumors (435/Neo.luc) during the same time interval (ANOVA, p = 0.005). The principal differences after 110 days were found in bones, which had 1.5 × 105 ± 1.2 × 105 tumor cell equivalents (p = 0.03), and lymph nodes, which had 7.0 × 106 ± 6.0 × 106 tumor cell equivalents (p = 0.08). The analyses of light production by tissues at different times showed that cells from 435/Neo.luc and 435/Bcl-xL.luc tumors were detectable in several organs by the second day after intramammary fat pad implantation. Although initially arriving at the target organs in similar numbers, 435/Bcl-xL cells developed more metastases than 435/Neo cells, indicating that the Bcl-xL gene might have a role in breast cancer dormancy, promoting survival of cells in metastatic foci. Thus, we suggest that overexpression of Bcl-xL could counteract the proapoptotic signals in the microenvironment and favor the successful development of metastasis in specific organs.


The Prostate | 2000

Metastatic burden in nude mice organs measured using prostate tumor PC-3 cells expressing the luciferase gene as a quantifiable tumor cell marker.

Nuria Rubio; Mercedes Martinez Villacampa; Nadia El Hilali; Jerónimo Blanco

Sensitive procedures for quantitative measurement of tumor cell spread as a function of time and primary tumor size are necessary to generate models of metastasis and formulate therapies.


Biomaterials | 2009

Dual luciferase labelling for non-invasive bioluminescence imaging of mesenchymal stromal cell chondrogenic differentiation in demineralized bone matrix scaffolds

Marta Vilalta; Christian Jorgensen; Irene R. Dégano; Yuti Chernajovsky; David Gould; Danièle Noël; José A. Andrades; José Becerra; Nuria Rubio; Jerónimo Blanco

Non-invasive bioluminescence imaging (BLI) to monitor changes in gene expression of cells implanted in live animals should facilitate the development of biomaterial scaffolds for tissue regeneration. We show that, in vitro, induction of chondrogenic differentiation in mouse bone marrow stromal cell line (CL1) and human adipose tissue derived mesenchymal stromal cells (hAMSCs), permanently transduced with a procollagen II (COL2A1) promoter driving a firefly luciferase gene reporter (PLuc) (COL2A1p.PLuc), induces PLuc expression in correlation with increases in COL2A1 and Sox9 mRNA expression and acquisition of chondrocytic phenotype. To be able to simultaneously monitor in vivo cell differentiation and proliferation, COL2A1p.PLuc labelled cells were also genetically labelled with a renilla luciferase (RLuc) gene driven by a constitutively active cytomegalovirus promoter, and then seeded in demineralized bone matrix (DBM) subcutaneously implanted in SCID mice. Non-invasive BLI monitoring of the implanted mice showed that the PLuc/RLuc ratio reports on gene expression changes indicative of cell differentiation. Large (CL1) and moderated (hAMSCs) changes in the PLuc/RLuc ratio over a 6 week period, revealed different patterns of in vivo chondrogenic differentiation for the CL1 cell line and primary MSCs, in agreement with in vitro published data and our results from histological analysis of DBM sections. This double bioluminescence labelling strategy together with BLI imaging to analyze behaviour of cells implanted in live animals should facilitate the development of progenitor cell/scaffold combinations for tissue repair.


International Journal of Cancer | 2009

HER3 is required for the maintenance of neuregulin‐dependent and ‐independent attributes of malignant progression in prostate cancer cells

Marta Soler; Francesca Mancini; Óscar Meca-Cortés; Lourdes Sánchez-Cid; Nuria Rubio; Sonia Lopez-Fernandez; Juan José Lozano; Jerónimo Blanco; Pedro L. Fernández; Timothy M. Thomson

HER3 (ERBB3) is a catalytically inactive pseudokinase of the HER receptor tyrosine kinase family, frequently overexpressed in prostate and other cancers. Aberrant expression and mutations of 2 other members of the family, EGFR and HER2, are key carcinogenic events in several types of tumors, and both are well‐ validated therapeutic targets. In this study, we show that HER3 is required to maintain the motile and invasive phenotypes of prostate (DU‐145) and breast (MCF‐7) cancer cells in response to the HER3 ligand neuregulin‐1 (NRG‐1), epidermal growth factor (EGF) and fetal bovine serum. Although MCF‐7 breast cancer cells appeared to require HER3 as part of an autocrine response induced by EGF and FBS, the response of DU‐145 prostate cancer cells to these stimuli, while requiring HER3, did not appear to involve autocrine stimulation of the receptor. DU‐145 cells required the expression of HER3 for efficient clonogenicity in vitro in standard growth medium and for tumorigenicity in immunodeficient mice. These observations suggest that prostate cancer cells derived from tumors that overexpress HER3 are dependent on its expression for the maintenance of major attributes of neoplastic aggressiveness, with or without cognate ligand stimulation.


Biomaterials | 2009

The effect of self-assembling peptide nanofiber scaffolds on mouse embryonic fibroblast implantation and proliferation.

Irene R. Dégano; Lluís Quintana; Marta Vilalta; David Horna; Nuria Rubio; Salvador Borrós; Carlos E. Semino; Jerónimo Blanco

Development of new materials for tissue engineering can be facilitated by the capacity to efficiently monitor in vivo the survival, proliferation and differentiation behaviour of cells implanted in different target tissues. We present here the application of a previously developed platform that allows to monitor in real time the survival and proliferative behaviour of implanted cells in two anatomical sites: subcutaneous and intramuscular. Basically, the system is based on the use of a non-invasive bioluminescence imaging (BLI) technique to detect luciferase expressing C57BL/6 cells, mouse embryonic fibroblasts, seeded in two sets of scaffolds: 1, a RAD16-I self-assembling peptide nanofiber matrix and 2, a composite consisted of the same RAD16-I nanofibers contained into a microporous biorubber scaffold. Interestingly, our results indicated considerable differences in the behaviour of implanted cells in each scaffold type. We observed that the self-assembling peptide scaffold alone foster cell survival and promotes cell proliferation where the composite scaffold not. Since self-assembling peptide scaffolds presents value stiffness proximal to the implanted tissues it is suggestive to think that harder materials will provide a physical constriction for cells to proliferate as well as mechanical discontinuity. We therefore propose that it is important to close match the implantation environment with the cell/material constructs in order to obtain the best response of the cells, illustrating the convenience of this strategy for the development of new tissue engineering platforms.


Gene Therapy | 2009

Human adipose tissue-derived mesenchymal stromal cells as vehicles for tumor bystander effect: a model based on bioluminescence imaging.

Marta Vilalta; Irene R. Dégano; Juli R. Bagó; Elisabet Aguilar; Sanjiv S. Gambhir; Nuria Rubio; Jerónimo Blanco

Human adipose tissue mesenchymal stromal cells (AMSCs) share common traits, including similar differentiation potential and cell surface markers, with their bone marrow counterparts. Owing to their general availability, higher abundance and ease of isolation AMSCs may be convenient autologous delivery vehicles for localized tumor therapy. We demonstrate a model for tumor therapy development based on the use of AMSCs expressing renilla luciferase and thymidine kinase, as cellular vehicles for ganciclovir-mediated bystander killing of firefly luciferase expressing tumors, and noninvasive bioluminescence imaging to continuously monitor both, tumor cells and AMSCs. We show that the therapy delivering AMSCs survive long time within tumors, optimize the ratio of AMSCs to tumor cells for therapy, and asses the therapeutic effect in real time. Treatment of mice bearing prostate tumors plus therapeutic AMSCs with the prodrug ganciclovir induced bystander killing effect, reducing the number of tumor cells to 1.5 % that of control tumors. Thus, AMSCs could be useful vehicles to deliver localized therapy, with potential for clinical application in inoperable tumors and surgical borders after tumor resection. This approach, useful to evaluate efficiency of therapeutic models, should facilitate the selection of cell types, dosages, therapeutic agents and treatment protocols for cell-based therapies of specific tumors.

Collaboration


Dive into the Jerónimo Blanco's collaboration.

Top Co-Authors

Avatar

Marta Guerra-Rebollo

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Olaia F. Vila

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Timothy M. Thomson

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Carolina Soler-Botija

Instituto de Salud Carlos III

View shared research outputs
Researchain Logo
Decentralizing Knowledge