Network


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

Hotspot


Dive into the research topics where Vanesa Santos is active.

Publication


Featured researches published by Vanesa Santos.


Cancer Research | 2008

Lysyl Oxidase-Like 2 as a New Poor Prognosis Marker of Squamous Cell Carcinomas

Héctor Peinado; Gema Moreno-Bueno; David Hardisson; Eduardo Pérez-Gómez; Vanesa Santos; Marta Mendiola; Juan Ignacio de Diego; Manuel Nistal; Miguel Quintanilla; Francisco Portillo; Amparo Cano

Lysyl oxidase-like 2 (Loxl2) interacts with and stabilizes Snai1 transcription factor, promoting epithelial-mesenchymal transition. Either Loxl2 or Snai1 knock-down blocks tumor growth and induces differentiation, but the specific role of each factor in tumor progression is still unknown. Comparison of the gene expression profiles of the squamous cell carcinoma cell line HaCa4 after knocking-down Loxl2 or Snai1 revealed that a subset of epidermal differentiation genes was specifically up-regulated in Loxl2-silenced cells. In agreement, although both Loxl2- and Snai1-knockdown cells showed reduced in vivo invasion, only Loxl2-silenced cells exhibited a skin-like epidermal differentiation program. In addition, we show that expression of Loxl2 and Snai1 correlates with malignant progression in a two-stage mouse skin carcinogenesis model. Furthermore, we found that increased expression of both LOXL2 and SNAI1 correlates with local recurrence in a cohort of 256 human laryngeal squamous cell carcinomas. We describe for the first time that high levels of LOXL2 are associated with decreased overall and disease-free survival in laryngeal squamous cell carcinomas, lung squamous cell carcinoma, and lymph node-negative (N(0)) breast adenocarcinomas. Altogether, our results show that LOXL2 can be used as a new poor prognosis indicator in human squamous cell carcinomas promoting malignant transformation by both SNAI1-dependent and SNAI1-independent pathways.


Nature Protocols | 2009

The morphological and molecular features of the epithelial-to-mesenchymal transition.

Gema Moreno-Bueno; Héctor Peinado; Patricia Molina; David Olmeda; Eva Cubillo; Vanesa Santos; José Palacios; Francisco Portillo; Amparo Cano

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.


Embo Molecular Medicine | 2011

Lysyl oxidase-like 2 (LOXL2), a new regulator of cell polarity required for metastatic dissemination of basal-like breast carcinomas

Gema Moreno-Bueno; Fernando Salvador; Alberto Martín; Alfredo Floristán; Eva P. Cuevas; Vanesa Santos; Amalia Montes; Saleta Morales; María Ángeles Castilla; Alejandro Rojo-Sebastián; Alejandra Nacarino Martínez; David Hardisson; Katalin Csiszar; Francisco Portillo; Héctor Peinado; José Palacios; Amparo Cano

Basal‐like breast carcinoma is characterized by the expression of basal/myoepithelial markers, undifferentiated phenotype, highly aggressive behaviour and frequent triple negative status (ESR−, PR−, Her2neu−). We have previously shown that epithelial–mesenchymal transition (EMT) occurs in basal‐like breast tumours and identified Lysyl‐oxidase‐like 2 (LOXL2) as an EMT player and poor prognosis marker in squamous cell carcinomas. We now show that LOXL2 mRNA is overexpressed in basal‐like human breast carcinomas. Breast carcinoma cell lines with basal‐like phenotype show a specific cytoplasmic/perinuclear LOXL2 expression, and this subcellular distribution is significantly associated with distant metastatic incidence in basal‐like breast carcinomas. LOXL2 silencing in basal‐like carcinoma cells induces a mesenchymal‐epithelial transition (MET) associated with a decrease of tumourigenicity and suppression of metastatic potential. Mechanistic studies indicate that LOXL2 maintains the mesenchymal phenotype of basal‐like carcinoma cells by a novel mechanism involving transcriptional downregulation of Lgl2 and claudin1 and disorganization of cell polarity and tight junction complexes. Therefore, intracellular LOXL2 is a new candidate marker of basal‐like carcinomas and a target to block metastatic dissemination of this aggressive breast tumour subtype.


PLOS ONE | 2012

Characterization of the SNAG and SLUG domains of Snail2 in the repression of E-cadherin and EMT induction: modulation by serine 4 phosphorylation.

Patricia Molina-Ortiz; Ana Villarejo; Matthew MacPherson; Vanesa Santos; Amalia Montes; Serhiy Souchelnytskyi; Francisco Portillo; Amparo Cano

Snail1 and Snail2, two highly related members of the Snail superfamily, are direct transcriptional repressors of E-cadherin and EMT inducers. Previous comparative gene profiling analyses have revealed important differences in the gene expression pattern regulated by Snail1 and Snail2, indicating functional differences between both factors. The molecular mechanism of Snail1-mediated repression has been elucidated to some extent, but very little is presently known on the repression mediated by Snail2. In the present work, we report on the characterization of Snail2 repression of E-cadherin and its regulation by phosphorylation. Both the N-terminal SNAG and the central SLUG domains of Snail2 are required for efficient repression of the E-cadherin promoter. The co-repressor NCoR interacts with Snail2 through the SNAG domain, while CtBP1 is recruited through the SLUG domain. Interestingly, the SNAG domain is absolutely required for EMT induction while the SLUG domain plays a negative modulation of Snail2 mediated EMT. Additionally, we identify here novel in vivo phosphorylation sites at serine 4 and serine 88 of Snail2 and demonstrate the functional implication of serine 4 in the regulation of Snail2-mediated repressor activity of E-cadherin and in Snail2 induction of EMT.


Oncogene | 2015

Lysyl oxidase-like 2 (LOXL2) and E47 EMT factor: novel partners in E-cadherin repression and early metastasis colonization

Giacomo Canesin; Eva P. Cuevas; Vanesa Santos; Celia López-Menéndez; Gema Moreno-Bueno; Yujie Huang; Katalin Csiszar; Francisco Portillo; Héctor Peinado; David Lyden; Amparo Cano

Epithelial–mesenchymal transition (EMT) has been associated with increased aggressiveness and acquisition of migratory properties providing tumor cells with the ability to invade into adjacent tissues. Downregulation of E-cadherin, a hallmark of EMT, is mediated by several transcription factors (EMT-TFs) that act also as EMT inducers, among them, Snail1 and the bHLH transcription factor E47. We previously described lysyl oxidase-like 2 (LOXL2), a member of the lysyl oxidase family, as a Snail1 regulator and EMT inducer. Here we show that LOXL2 is also an E47-interacting partner and functionally collaborates in the repression of E-cadherin promoter. Loss and gain of function analyses combined with in vivo studies in syngeneic breast cancer models demonstrate the participation of LOXL2 and E47 in tumor growth and their requirement for lung metastasis. Furthermore, LOXL2 and E47 contribute to early steps of metastatic colonization by cell and noncell autonomous functions regulating the recruitment of bone marrow progenitor cells to the lungs and by direct transcriptional regulation of fibronectin and cytokines TNFα, ANG-1 and GM-CSF. Moreover, fibronectin and GM-CSF proved to be necessary for LOXL2/E47-mediated modulation of tumor growth and lung metastasis.


PLOS ONE | 2013

E47 and Id1 Interplay in Epithelial-Mesenchymal Transition

Eva Cubillo; Antonio Díaz-López; Eva P. Cuevas; Gema Moreno-Bueno; Héctor Peinado; Amalia Montes; Vanesa Santos; Francisco Portillo; Amparo Cano

E12/E47 proteins (encoded by E2A gene) are members of the class I basic helix-loop-helix (bHLH) transcription factors (also known as E proteins). E47 has been described as repressor of E-cadherin and inducer of epithelial-mesenchymal transition (EMT). We reported previously that EMT mediated by E47 in MDCK cells occurs with a concomitant overexpression of Id1 and Id3 proteins. Id proteins belong to class V of HLH factors that lack the basic domain; they dimerise with E proteins and prevent their DNA interaction, thus, acting as dominant negative of E proteins. Here, we show that E47 interacts with Id1 in E47 overexpressing MDCK cells that underwent a full EMT as well as in mesenchymal breast carcinoma and melanoma cell lines. By conducting chromatin immunoprecipitation assays we demonstrate that E47 binds directly to the endogenous E-cadherin promoter of mesenchymal MDCK-E47 cells in a complex devoid of Id1. Importantly, our data suggest that both E47 and Id1 are required to maintain the mesenchymal phenotype of MDCK-E47 cells. These data support the collaboration between E47 and Id1 in the maintenance of EMT by mechanisms independent of the dominant negative action of Id1 on E47 binding to E-cadherin promoter. Finally, the analysis of several N0 breast tumour series indicates that the expression of E47 and ID1 is significantly associated with the basal-like phenotype supporting the biological significance of the present findings.


Biology Open | 2014

LOXL2 catalytically inactive mutants mediate epithelial-to-mesenchymal transition

Eva P. Cuevas; Gema Moreno-Bueno; Giacomo Canesin; Vanesa Santos; Francisco Portillo; Amparo Cano

Summary Lysyl-oxidase-like 2 (LOXL2) is a member of the lysyl oxidase family that catalyzes the cross-linking of collagens or elastins in the extracellular matrix, thus regulating the tensile strength of tissues. However, many reports have suggested different intracellular roles for LOXL2, including the ability to regulate gene transcription and tumor progression. We previously reported that LOXL2 mediates epithelial-to-mesenchymal transition (EMT) by Snail1-dependent and independent mechanisms, related to E-cadherin silencing and downregulation of epidermal differentiation and cell polarity components, respectively. Whether or not the catalytic activity of LOXL2 is required to induce/sustain EMT is actually unknown. Here we show that LOXL2 catalytic inactive mutants collaborate with Snail1 in E-cadherin gene repression to trigger EMT and, in addition, promote FAK/Src pathway activation to support EMT. These findings reveal a non-conventional role of LOXL2 on regulating epithelial cell plasticity.


The EMBO Journal | 2015

Lysyl oxidase‐like 2 represses Notch1 expression in the skin to promote squamous cell carcinoma progression

Alberto Martín; Fernando Salvador; Gema Moreno-Bueno; Alfredo Floristán; Cristina Ruiz-Herguido; Eva P. Cuevas; Saleta Morales; Vanesa Santos; Katalin Csiszar; Pierre Dubus; Jody J. Haigh; Anna Bigas; Francisco Portillo; Amparo Cano

Lysyl oxidase‐like 2 (LOXL2) is involved in a wide range of physiological and pathological processes, including fibrosis and tumor progression, implicating intracellular and extracellular functions. To explore the specific in vivo role of LOXL2 in physiological and tumor contexts, we generated conditional gain‐ and loss‐of‐function mouse models. Germ‐line deletion of Loxl2 promotes lethality in half of newborn mice mainly associated to congenital heart defects, while Loxl2 overexpression triggers male sterility due to epididymal dysfunction caused by epithelial disorganization, fibrosis and acute inflammation. Remarkably, when challenged to chemical skin carcinogenesis, Loxl2‐overexpressing mice increased tumor burden and malignant progression, while Loxl2‐deficient mice exhibit the opposite phenotypes. Loxl2 levels in premalignant tumors negatively correlate with expression of epidermal differentiation markers and components of the Notch1 pathway. We show that LOXL2 is a direct repressor of NOTCH1. Additionally, we identify an exclusive expression pattern between LOXL2 and members of the canonical NOTCH1 pathway in human HNSCC. Our data identify for the first time novel LOXL2 roles in tissue homeostasis and support it as a target for SCC therapy.


International Journal of Cancer | 2015

Zeb1 and Snail1 engage miR-200f transcriptional and epigenetic regulation during EMT

Antonio Díaz-López; Juan Díaz-Martín; Gema Moreno-Bueno; Eva P. Cuevas; Vanesa Santos; David Olmeda; Francisco Portillo; José Palacios; Amparo Cano

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.


Scientific Reports | 2017

LOXL2 drives epithelial-mesenchymal transition via activation of IRE1-XBP1 signalling pathway

Eva P. Cuevas; Pilar Eraso; María J. Mazón; Vanesa Santos; Gema Moreno-Bueno; Amparo Cano; Francisco Portillo

Epithelial-to-Mesenchymal Transition (EMT) is a key process contributing to the aggressiveness of cancer cells. EMT is triggered by activation of different transcription factors collectively known as EMT-TFs. Different cellular cues and cell signalling networks activate EMT at transcriptional and posttranscriptional level in different biological and pathological situations. Among them, overexpression of LOXL2 (lysyl oxidase-like 2) induces EMT independent of its catalytic activity. Remarkably, perinuclear/cytoplasmic accumulation of LOXL2 is a poor prognosis marker of squamous cell carcinomas and is associated to basal breast cancer metastasis by mechanisms no yet fully understood. Here, we report that overexpression of LOXL2 promotes its accumulation in the Endoplasmic Reticulum where it interacts with HSPA5 leading to activation of the IRE1-XBP1 signalling pathway of the ER-stress response. LOXL2-dependent IRE1-XBP1 activation induces the expression of several EMT-TFs: SNAI1, SNAI2, ZEB2 and TCF3 that are direct transcriptional targets of XBP1. Remarkably, inhibition of IRE1 blocks LOXL2-dependent upregulation of EMT-TFs thus hindering EMT induction.

Collaboration


Dive into the Vanesa Santos's collaboration.

Top Co-Authors

Avatar

Amparo Cano

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Francisco Portillo

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Gema Moreno-Bueno

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Eva P. Cuevas

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saleta Morales

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Fernando Salvador

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Alberto Martín

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Alfredo Floristán

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Amalia Montes

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge