Network


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

Hotspot


Dive into the research topics where Gustavo Rodríguez-Esteban is active.

Publication


Featured researches published by Gustavo Rodríguez-Esteban.


The International Journal of Developmental Biology | 2009

Planarian regeneration: achievements and future directions after 20 years of research

Emili Saló; Josep F. Abril; Teresa Adell; Francesc Cebrià; Kay Eckelt; Enrique Fernández-Taboada; Mette Handberg-Thorsager; Marta Iglesias; M. Dolores Molina; Gustavo Rodríguez-Esteban

Planarians can undergo dramatic changes in body size and regenerate their entire body plan from small pieces after cutting. This remarkable morphological plasticity has made them an excellent model in which to analyze phenomena such as morphogenesis, restoration of pattern and polarity, control of tissue proportions and tissue homeostasis. They have a unique population of pluripotent stem cells in the adult that can give rise to all differentiated cell types, including the germ cells. These cellular characteristics provide an excellent opportunity to study the mechanisms involved in the maintenance and differentiation of cell populations in intact and regenerating animals. Until recently, the planarian model system lacked opportunities for genetic analysis; however, this handicap was overcome in the last decade through the development of new molecular methods which have been successfully applied to planarians. These techniques have allowed analysis of the temporal and spatial expression of genes, as well as interference with gene function, generating the first phenotypes by loss or gain of function. Finally, the sequencing of the planarian genome has provided the essential tools for an in-depth analysis of the genomic regulation of this model system. In this review, we provide an overview of planarians as a model system for research into development and regeneration and describe new lines of investigation in this area.


BMC Genomics | 2010

Smed454 dataset: unravelling the transcriptome of Schmidtea mediterranea

Josep F. Abril; Francesc Cebrià; Gustavo Rodríguez-Esteban; Thomas Horn; Susanna Fraguas; Beatriz Calvo; Kerstin Bartscherer; Emili Saló

BackgroundFreshwater planarians are an attractive model for regeneration and stem cell research and have become a promising tool in the field of regenerative medicine. With the availability of a sequenced planarian genome, the recent application of modern genetic and high-throughput tools has resulted in revitalized interest in these animals, long known for their amazing regenerative capabilities, which enable them to regrow even a new head after decapitation. However, a detailed description of the planarian transcriptome is essential for future investigation into regenerative processes using planarians as a model system.ResultsIn order to complement and improve existing gene annotations, we used a 454 pyrosequencing approach to analyze the transcriptome of the planarian species Schmidtea mediterranea Altogether, 598,435 454-sequencing reads, with an average length of 327 bp, were assembled together with the ~10,000 sequences of the S. mediterranea UniGene set using different similarity cutoffs. The assembly was then mapped onto the current genome data. Remarkably, our Smed454 dataset contains more than 3 million novel transcribed nucleotides sequenced for the first time. A descriptive analysis of planarian splice sites was conducted on those Smed454 contigs that mapped univocally to the current genome assembly. Sequence analysis allowed us to identify genes encoding putative proteins with defined structural properties, such as transmembrane domains. Moreover, we annotated the Smed454 dataset using Gene Ontology, and identified putative homologues of several gene families that may play a key role during regeneration, such as neurotransmitter and hormone receptors, homeobox-containing genes, and genes related to eye function.ConclusionsWe report the first planarian transcript dataset, Smed454, as an open resource tool that can be accessed via a web interface. Smed454 contains significant novel sequence information about most expressed genes of S. mediterranea. Analysis of the annotated data promises to contribute to identification of gene families poorly characterized at a functional level. The Smed454 transcriptome data will assist in the molecular characterization of S. mediterranea as a model organism, which will be useful to a broad scientific community.


The International Journal of Developmental Biology | 2012

Decreased neoblast progeny and increased cell death during starvation-induced planarian degrowth

Cristina González-Estévez; Daniel A. Felix; Gustavo Rodríguez-Esteban; A. Aziz Aboobaker

The development of a complex multicellular organism requires a careful coordination of growth, cell division, cell differentiation and cell death. All these processes must be under intricate and coordinated control, as they have to be integrated across all tissues. Freshwater planarians are especially plastic, in that they constantly replace somatic tissues from a pool of adult somatic stem cells and continuously undergo growth and degrowth as adult animals in response to nutrient availability. During these processes they appear to maintain perfect scale of tissues and organs. These life history traits make them an ideal model system to study growth and degrowth. We have studied the unique planarian process of degrowth. When food is not available, planarians are able to degrow to a minimum size, without any signs of adverse physiological outcomes. For example they maintain full regenerative capacity. Our current knowledge of how this is regulated at the molecular and cellular level is very limited. Planarian degrowth has been reported to result from a decrease in cell number rather than a decrease in cell size. Thus one obvious explanation for degrowth would be a decrease in stem cell proliferation. However evidence in the literature suggests this is not the case. We show that planarians maintain normal basal mitotic rates during degrowth but that the number of stem cell progeny decreases during starvation and degrowth. These observations are reversed upon feeding, indicating that they are dependent on nutritional status. An increase in cell death is also observed during degrowth, which is not rapidly reversed upon feeding. We conclude that degrowth is a result of cell death decreasing cell numbers and that the dynamics of neoblast self-renewal and differentiation adapt to nutrient conditions to allow maintenance of the neoblast population during the period of starvation.


BMC Genomics | 2011

A proteomics approach to decipher the molecular nature of planarian stem cells

Enrique Fernández-Taboada; Gustavo Rodríguez-Esteban; Emili Saló; Josep F. Abril

BackgroundIn recent years, planaria have emerged as an important model system for research into stem cells and regeneration. Attention is focused on their unique stem cells, the neoblasts, which can differentiate into any cell type present in the adult organism. Sequencing of the Schmidtea mediterranea genome and some expressed sequence tag projects have generated extensive data on the genetic profile of these cells. However, little information is available on their protein dynamics.ResultsWe developed a proteomic strategy to identify neoblast-specific proteins. Here we describe the method and discuss the results in comparison to the genomic high-throughput analyses carried out in planaria and to proteomic studies using other stem cell systems. We also show functional data for some of the candidate genes selected in our proteomic approach.ConclusionsWe have developed an accurate and reliable mass-spectra-based proteomics approach to complement previous genomic studies and to further achieve a more accurate understanding and description of the molecular and cellular processes related to the neoblasts.


Development | 2014

egr-4, a target of EGFR signaling, is required for the formation of the brain primordia and head regeneration in planarians

Susanna Fraguas; Sara Barberán; Marta Iglesias; Gustavo Rodríguez-Esteban; Francesc Cebrià

During the regeneration of freshwater planarians, polarity and patterning programs play essential roles in determining whether a head or a tail regenerates at anterior or posterior-facing wounds. This decision is made very soon after amputation. The pivotal role of the Wnt/β-catenin and Hh signaling pathways in re-establishing anterior-posterior (AP) polarity has been well documented. However, the mechanisms that control the growth and differentiation of the blastema in accordance with its AP identity are less well understood. Previous studies have described a role of Smed-egfr-3, a planarian epidermal growth factor receptor, in blastema growth and differentiation. Here, we identify Smed-egr-4, a zinc-finger transcription factor belonging to the early growth response gene family, as a putative downstream target of Smed-egfr-3. Smed-egr-4 is mainly expressed in the central nervous system and its silencing inhibits anterior regeneration without affecting the regeneration of posterior regions. Single and combinatorial RNA interference to target different elements of the Wnt/β-catenin pathway, together with expression analysis of brain- and anterior-specific markers, revealed that Smed-egr-4: (1) is expressed in two phases – an early Smed-egfr-3-independent phase and a late Smed-egfr-3-dependent phase; (2) is necessary for the differentiation of the brain primordia in the early stages of regeneration; and (3) that it appears to antagonize the activity of the Wnt/β-catenin pathway to allow head regeneration. These results suggest that a conserved EGFR/egr pathway plays an important role in cell differentiation during planarian regeneration and indicate an association between early brain differentiation and the proper progression of head regeneration.


The International Journal of Developmental Biology | 2012

Heterogeneity of planarian stem cells in the S/G2/M phase.

Sören Moritz; Franziska Stöckle; Claudia Ortmeier; Henning Schmitz; Gustavo Rodríguez-Esteban; Göran Key; Luca Gentile

The planarian adult stem cell (pASC) population has a specific molecular signature and can be easily visualized and isolated by flow cytometry. However, the lack of antibodies against specific surface markers for planarian cells prevents a deeper analysis of specific cell populations. Here, if we describe the results of the immunoscreening of pASC plasma membrane proteins (PMPs). A novel papain-based method for planarian cell dissociation enabling both high yield and improved cell viability was used to generate single cell preparations for PMP purification. PMPs were used for intraperitoneal immunization of mice and thus about 1000 hybridoma clones were generated and screened. Supernatants collected from the hybridoma clones were first screened by ELISA and then by live immuno-staining. About half of these supernatants stained all the planarian cells, whereas the other half specifically labeled a subfraction thereof. A detailed analysis of two hybridoma supernatants revealed that large subfractions of the X1, X2 and Xin populations differentially express specific membrane markers. Quantitative PCR data disclosed a correlation between the immunostaining results and the expression of markers of the early and late progeny, also for those pASCs in the S/G2/M phase of the cell cycle (X1 population). Thus, about two thirds of the cycling pASCs showed a specific membrane signature coupled with the expression of markers hitherto considered to be restricted to differentiating, post-mitotic progeny. In summary, a library of 66 monoclonal antibodies against planarian PMPs was generated. The analysis of two of the clones generated revealed that a subset of cells of the X1 population expresses early and late progeny markers, which might indicate that these cells are committed while still proliferating. The findings demonstrate the usefulness of our PMP antibody library for planarian research.


BMC Genomics | 2015

Digital gene expression approach over multiple RNA-Seq data sets to detect neoblast transcriptional changes in Schmidtea mediterranea.

Gustavo Rodríguez-Esteban; Alejandro González-Sastre; Jose Ignacio Rojo-Laguna; Emili Saló; Josep F. Abril

BackgroundThe freshwater planarian Schmidtea mediterranea is recognised as a valuable model for research into adult stem cells and regeneration.With the advent of the high-throughput sequencing technologies, it has become feasible to undertake detailed transcriptional analysis of its unique stem cell population, the neoblasts. Nonetheless, a reliable reference for this type of studies is still lacking.ResultsTaking advantage of digital gene expression (DGE) sequencing technology we compare all the available transcriptomes for S. mediterranea and improve their annotation. These results are accessible via web for the community of researchers.Using the quantitative nature of DGE, we describe the transcriptional profile of neoblasts and present 42 new neoblast genes, including several cancer-related genes and transcription factors. Furthermore, we describe in detail the Smed-meis-like gene and the three Nuclear Factor Y subunits Smed-nf-YA, Smed-nf-YB-2 and Smed-nf-YC.ConclusionsDGE is a valuable tool for gene discovery, quantification and annotation. The application of DGE in S. mediterranea confirms the planarian stem cells or neoblasts as a complex population of pluripotent and multipotent cells regulated by a mixture of transcription factors and cancer-related genes.


Annals of Oncology | 2017

Dual MET and ERBB inhibition overcomes intratumor plasticity in osimertinib-resistant-advanced non-small-cell lung cancer (NSCLC)

Alex Martinez-Marti; Enriqueta Felip; J. Matito; Elisabetta Mereu; Alejandro Navarro; S. Cedres; Nuria Pardo; A. Martinez de Castro; J Remon; Josep M Miquel; Amy Guillaumet-Adkins; E. Nadal; Gustavo Rodríguez-Esteban; O. Arqués; R. Fasani; Paolo Nuciforo; Holger Heyn; Alberto Villanueva; H. G. Palmer; Ana Vivancos

Abstract Background Third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) such as osimertinib are the last line of targeted treatment of metastatic non-small-cell lung cancer (NSCLC) EGFR-mutant harboring T790M. Different mechanisms of acquired resistance to third-generation EGFR-TKIs have been proposed. It is therefore crucial to identify new and effective strategies to overcome successive acquired mechanisms of resistance. Methods For Amplicon-seq analysis, samples from the index patient (primary and metastasis lesions at different timepoints) as well as the patient-derived orthotopic xenograft tumors corresponding to the different treatment arms were used. All samples were formalin-fixed paraffin-embedded, selected and evaluated by a pathologist. For droplet digital PCR, 20 patients diagnosed with NSCLC at baseline or progression to different lines of TKI therapies were selected. Formalin-fixed paraffin-embedded blocks corresponding to either primary tumor or metastasis specimens were used for analysis. For single-cell analysis, orthotopically grown metastases were dissected from the brain of an athymic nu/nu mouse and cryopreserved at −80°C. Results In a brain metastasis lesion from a NSCLC patient presenting an EGFR T790M mutation, we detected MET gene amplification after prolonged treatment with osimertinib. Importantly, the combination of capmatinib (c-MET inhibitor) and afatinib (ErbB-1/2/4 inhibitor) completely suppressed tumor growth in mice orthotopically injected with cells derived from this brain metastasis. In those mice treated with capmatinib or afatinib as monotherapy, we observed the emergence of KRAS G12C clones. Single-cell gene expression analyses also revealed intratumor heterogeneity, indicating the presence of a KRAS-driven subclone. We also detected low-frequent KRAS G12C alleles in patients treated with various EGFR-TKIs. Conclusion Acquired resistance to subsequent EGFR-TKI treatment lines in EGFR-mutant lung cancer patients may induce genetic plasticity. We assess the biological insights of tumor heterogeneity in an osimertinib-resistant tumor with acquired MET-amplification and propose new treatment strategies in this situation.


PLOS Biology | 2018

Hippo signaling controls cell cycle and restricts cell plasticity in planarians.

Nídia Sousa; Gustavo Rodríguez-Esteban; Jose Ignacio Rojo-Laguna; Emili Saló; Teresa Adell

The Hippo pathway plays a key role in regulating cell turnover in adult tissues, and abnormalities in this pathway are consistently associated with human cancers. Hippo was initially implicated in the control of cell proliferation and death, and its inhibition is linked to the expansion of stem cells and progenitors, leading to larger organ size and tumor formation. To understand the mechanism by which Hippo directs cell renewal and promotes stemness, we studied its function in planarians. These stem cell–based organisms are ideal models for the analysis of the complex cellular events underlying tissue renewal in the whole organism. hippo RNA interference (RNAi) in planarians decreased apoptotic cell death, induced cell cycle arrest, and could promote the dedifferentiation of postmitotic cells. hippo RNAi resulted in extensive undifferentiated areas and overgrowths, with no effect on body size or cell number. We propose an essential role for hippo in controlling cell cycle, restricting cell plasticity, and thereby preventing tumoral transformation.


Genome Research | 2018

Bigscale: An analytical framework for big-scale single-cell data

Giovanni Iacono; Elisabetta Mereu; Amy Guillaumet-Adkins; Roser Corominas; Ivon Cuscó; Gustavo Rodríguez-Esteban; Marta Gut; Luis A. Pérez-Jurado; Ivo Gut; Holger Heyn

Single-cell RNA sequencing (scRNA-seq) has significantly deepened our insights into complex tissues, with the latest techniques capable of processing tens of thousands of cells simultaneously. Analyzing increasing numbers of cells, however, generates extremely large data sets, extending processing time and challenging computing resources. Current scRNA-seq analysis tools are not designed to interrogate large data sets and often lack sensitivity to identify marker genes. With bigSCale, we provide a scalable analytical framework to analyze millions of cells, which addresses the challenges associated with large data sets. To handle the noise and sparsity of scRNA-seq data, bigSCale uses large sample sizes to estimate an accurate numerical model of noise. The framework further includes modules for differential expression analysis, cell clustering, and marker identification. A directed convolution strategy allows processing of extremely large data sets, while preserving transcript information from individual cells. We evaluated the performance of bigSCale using both a biological model of aberrant gene expression in patient-derived neuronal progenitor cells and simulated data sets, which underlines the speed and accuracy in differential expression analysis. To test its applicability for large data sets, we applied bigSCale to assess 1.3 million cells from the mouse developing forebrain. Its directed down-sampling strategy accumulates information from single cells into index cell transcriptomes, thereby defining cellular clusters with improved resolution. Accordingly, index cell clusters identified rare populations, such as reelin (Reln)-positive Cajal-Retzius neurons, for which we report previously unrecognized heterogeneity associated with distinct differentiation stages, spatial organization, and cellular function. Together, bigSCale presents a solution to address future challenges of large single-cell data sets.

Collaboration


Dive into the Gustavo Rodríguez-Esteban's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emili Saló

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Holger Heyn

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar

Ivo Gut

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marta Gut

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alex Martinez-Marti

Autonomous University of Barcelona

View shared research outputs
Researchain Logo
Decentralizing Knowledge