Network


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

Hotspot


Dive into the research topics where Amaia Vilas-Zornoza is active.

Publication


Featured researches published by Amaia Vilas-Zornoza.


Cancer Research | 2009

Epigenetic Silencing of the Tumor Suppressor MicroRNA Hsa-miR-124a Regulates CDK6 Expression and Confers a Poor Prognosis in Acute Lymphoblastic Leukemia

Xabier Agirre; Amaia Vilas-Zornoza; Antonio Jiménez-Velasco; José I. Martín-Subero; Lucia Cordeu; Leire Garate; Edurne San José-Enériz; Gloria Abizanda; Paula Rodriguez-Otero; Puri Fortes; José Rifón; Eva Bandrés; María José Calasanz; Vanesa Martín; Anabel Heiniger; Antonio Torres; Reiner Siebert; Jose Roman-Gomez; Felipe Prosper

Whereas transcriptional silencing of genes due to epigenetic mechanisms is one of the most important alterations in acute lymphoblastic leukemia (ALL), some recent studies indicate that DNA methylation contributes to down-regulation of miRNAs during tumorigenesis. To explore the epigenetic alterations of miRNAs in ALL, we analyzed the methylation and chromatin status of the miR-124a loci in ALL. Expression of miR-124a was down-regulated in ALL by hypermethylation of the promoter and histone modifications including decreased levels of 3mk4H3 and AcH3 and increased levels of 2mK9H3, 3mK9H3, and 3mK27H3. Epigenetic down-regulation of miR-124a induced an up-regulation of its target, CDK6, and phosphorylation of retinoblastoma (Rb) and contributed to the abnormal proliferation of ALL cells both in vitro and in vivo. Cyclin-dependent kinase 6 (CDK6) inhibition by sodium butyrate or PD-0332991 decreased ALL cell growth in vitro, whereas overexpression of pre-miR124a led to decreased tumorigenicity in a xenogeneic in vivo Rag2(-/-)gammac(-/-) mouse model. The clinical implications of these findings were analyzed in a group of 353 patients diagnosed with ALL. Methylation of hsa-miR-124a was observed in 59% of the patients, which correlated with down-regulation of miR-124a (P < 0.001). Furthermore, hypermethylation of hsa-miR-124a was associated with higher relapse rate (P = 0.001) and mortality rate (P < 0.001), being an independent prognostic factor for disease-free survival (P < 0.001) and overall survival (P = 0.005) in the multivariate analysis. These results provide the grounds for new therapeutic strategies in ALL either targeting the epigenetic regulation of microRNAs and/or directly targeting the CDK6-Rb pathway.


Molecular Cancer Research | 2008

Down-Regulation of hsa-miR-10a in Chronic Myeloid Leukemia CD34+ Cells Increases USF2-Mediated Cell Growth

Xabier Agirre; Antonio Jiménez-Velasco; Edurne San José-Enériz; Leire Garate; Eva Bandrés; Lucia Cordeu; Oscar Aparicio; Borja Saez; German Navarro; Amaia Vilas-Zornoza; Ignacio Pérez-Roger; Jesús García-Foncillas; Antonio J. Torres; Anabel Heiniger; María José Calasanz; Puri Fortes; Jose Roman-Gomez; Felipe Prosper

MicroRNAs (miRNA) are small noncoding, single-stranded RNAs that inhibit gene expression at a posttranscriptional level, whose abnormal expression has been described in different tumors. The aim of our study was to identify miRNAs potentially implicated in chronic myeloid leukemia (CML). We detected an abnormal miRNA expression profile in mononuclear and CD34+ cells from patients with CML compared with healthy controls. Of 157 miRNAs tested, hsa-miR-10a, hsa-miR-150, and hsa-miR-151 were down-regulated, whereas hsa-miR-96 was up-regulated in CML cells. Down-regulation of hsa-miR-10a was not dependent on BCR-ABL1 activity and contributed to the increased cell growth of CML cells. We identified the upstream stimulatory factor 2 (USF2) as a potential target of hsa-miR-10a and showed that overexpression of USF2 also increases cell growth. The clinical relevance of these findings was shown in a group of 85 newly diagnosed patients with CML in which expression of hsa-miR-10a was down-regulated in 71% of the patients, whereas expression of USF2 was up-regulated in 60% of the CML patients, with overexpression of USF2 being significantly associated with decreased expression of hsa-miR-10a (P = 0.004). Our results indicate that down-regulation of hsa-miR-10a may increase USF2 and contribute to the increase in cell proliferation of CML implicating a miRNA in the abnormal behavior of CML. (Mol Cancer Res 2008;6(12):1830–40)


Molecular Cancer | 2009

MicroRNA expression profiling in Imatinib-resistant Chronic Myeloid Leukemia patients without clinically significant ABL1-mutations.

Edurne San José-Enériz; Jose Roman-Gomez; Antonio Jiménez-Velasco; Leire Garate; Vanesa Martin; Lucia Cordeu; Amaia Vilas-Zornoza; Paula Rodriguez-Otero; María José Calasanz; Felipe Prosper; Xabier Agirre

The development of Imatinib Mesylate (IM), the first specific inhibitor of BCR-ABL1, has had a major impact in patients with Chronic Myeloid Leukemia (CML), establishing IM as the standard therapy for CML. Despite the clinical success obtained with the use of IM, primary resistance to IM and molecular evidence of persistent disease has been observed in 20-25% of IM treated patients. The existence of second generation TK inhibitors, which are effective in patients with IM resistance, makes identification of predictors of resistance to IM an important goal in CML. In this study, we have identified a group of 19 miRNAs that may predict clinical resistance to IM in patients with newly diagnosed CML.


British Journal of Haematology | 2011

Deregulation of FGFR1 and CDK6 oncogenic pathways in acute lymphoblastic leukaemia harbouring epigenetic modifications of the MIR9 family.

Paula Rodriguez-Otero; Jose Roman-Gomez; Amaia Vilas-Zornoza; Edurne San José-Enériz; Vanesa Martín-Palanco; José Rifón; Antonio Torres; María José Calasanz; Xabier Agirre; Felipe Prosper

The role of epigenetic mechanisms in the regulation of microRNAs (miRNAs) with a tumour‐suppressor function in human neoplasms has recently been established. Several miRNAs have been found to be inappropriately regulated by DNA methylation in patients with acute lymphoblastic leukaemia (ALL). We analysed the methylation status of the three members of the MIR9 family (MIR9‐1, MIR9‐2 and MIR9‐3) in a uniformly treated cohort of 200 newly diagnosed ALLs. MIR9 was methylated in 54% of the patients and was associated with downregulation of MIR9 (P < 0·01). Hypermethylation of MIR9 was an independent prognostic factor for disease‐free survival, overall survival and event‐free survival in a multivariate analysis (P < 0·01). Epigenetic downregulation of MIR9 induced upregulation of its targets, FGFR1 and CDK6, while treatment of ALL cells with FGFR1 (PD‐173074) and CDK6 (PD‐0332991) inhibitors induced a decrease in cell proliferation and an increase in apoptosis of ALL cells. Our results indicate that the MIR9 family is involved in the pathogenesis and clinical behaviour of ALL and provide the basis for new therapeutic strategies in the treatment of ALL, targeting the epigenetic regulation of miRNAs and/or the FGFR1 or CDK6‐RB pathway directly.


Cancer Science | 2010

Epigenetic regulation of the non‐canonical Wnt pathway in acute myeloid leukemia

Vanesa Martin; Ana Valencia; Xabier Agirre; José Cervera; Edurne San José-Enériz; Amaia Vilas-Zornoza; Paula Rodriguez-Otero; Miguel A. Sanz; Concepción Herrera; Antonio J. Torres; Felipe Prosper; Jose Roman-Gomez

Wnt5a is a member of the Wnt family of proteins that signals through the non‐canonical Wnt/Ca2+pathway to suppress cyclin D1. Deregulation of this pathway has been found in animal models suggesting that it acts as tumour suppressor in acute myeloid leukemia (AML). Although DNA methylation is the main mechanism of regulation of the canonical Wnt pathway in AML, the role of WNT5A abnormalities has never been evaluated in this clinical setting. The methylation status of WNT5A promoter–exon 1 was analyzed by methylation‐specific PCR and sequencing in eleven AML‐derived cell lines and 252 AML patients. We observed WNT5A hypermethylation in seven cell lines and in 43% (107/252) of AML patients. WNT5A methylation was associated with decreased WNT5A expression (P < 0.001) that was restored after exposure to 5‐Aza‐2’‐deoxycytidine. Moreover, WNT5A hypermethylation correlated with upregulation of CYCLIN D1 expression (P < 0.001). Relapse (15%vs 37%, P < 0.001) and mortality (61%vs 79%, P = 0.004) rates were lower for patients in the non‐methylated group. Disease‐free survival and overall survival at 6 and 7 years, respectively, were 60% and 27% for unmethylated patients and 20% and 0% for hypermethylated patients (P = 0.0001 and P = 0.04, respectively). Interestingly, significant differences were also observed when the analysis was carried out according to cytogenetic risk groups. We demonstrate that WNT5A, a putative tumor suppressor gene in AML, is silenced by methylation in this disease and that this epigenetic event is associated with upregulation of CYCLIN D1 expression and confers poor prognosis in patients with AML. (Cancer Sci 2009)


Leukemia | 2012

Preclinical activity of LBH589 alone or in combination with chemotherapy in a xenogeneic mouse model of human acute lymphoblastic leukemia.

Amaia Vilas-Zornoza; Xabier Agirre; Gloria Abizanda; Cristina Moreno; Victor Segura; A De Martino Rodriguez; Edurne San José-Enériz; Estíbaliz Miranda; José I. Martín-Subero; Leire Garate; María J. Blanco-Prieto; J A García de Jalón; P Río; José Rifón; Juan C. Cigudosa; Jose A. Martinez-Climent; Jose Roman-Gomez; María José Calasanz; José Manuel Ribera; Felipe Prosper

Histone deacetylases (HDACs) have been identified as therapeutic targets due to their regulatory function in chromatin structure and organization. Here, we analyzed the therapeutic effect of LBH589, a class I–II HDAC inhibitor, in acute lymphoblastic leukemia (ALL). In vitro, LBH589 induced dose-dependent antiproliferative and apoptotic effects, which were associated with increased H3 and H4 histone acetylation. Intravenous administration of LBH589 in immunodeficient BALB/c-RAG2−/−γc−/− mice in which human-derived T and B-ALL cell lines were injected induced a significant reduction in tumor growth. Using primary ALL cells, a xenograft model of human leukemia in BALB/c-RAG2−/−γc−/− mice was established, allowing continuous passages of transplanted cells to several mouse generations. Treatment of mice engrafted with T or B-ALL cells with LBH589 induced an in vivo increase in the acetylation of H3 and H4, which was accompanied with prolonged survival of LBH589-treated mice in comparison with those receiving vincristine and dexamethasone. Notably, the therapeutic efficacy of LBH589 was significantly enhanced in combination with vincristine and dexamethasone. Our results show the therapeutic activity of LBH589 in combination with standard chemotherapy in pre-clinical models of ALL and suggest that this combination may be of clinical value in the treatment of patients with ALL.


Cancer Science | 2008

Methylation status of Wnt signaling pathway genes affects the clinical outcome of Philadelphia-positive acute lymphoblastic leukemia

Vanesa Martin; Xabier Agirre; Antonio Jiménez-Velasco; Edurne San José-Enériz; Lucia Cordeu; Leire Garate; Amaia Vilas-Zornoza; Juan A. Castillejo; Anabel Heiniger; Felipe Prosper; Antonio Torres; Jose Roman-Gomez

The clinical significance of aberrant promoter methylation of the canonical Wnt pathway antagonist genes (sFRP1, sFRP2, sFRP4, sFRP5, Wif1, Dkk3, and Hdpr1) and also putative tumor‐suppressor gene Wnt5a, belonging to the non‐canonical Wnt signaling pathway, was investigated in a large series of 75 patients with Philadelphia chromosome‐positive acute lymphoblastic leukemia by methylation‐specific polymerase chain reaction. At least one methylated gene was observed in cells from 66% (49/75) of patients (methylated group). Disease‐free survival and overall survival at 9 years were 51 and 40%, respectively, for the unmethylated group and 3 and 2%, respectively, for the methylated group (both P < 0.0001). Multivariate analysis demonstrated that the Wnt methylation profile was an independent prognostic factor predicting disease‐free survival (P = 0.007) and overall survival (P = 0.039). Abnormal DNA methylation of promoter‐associated CpG islands in the Wnt signaling pathway is very common in Philadelphia chromosome‐positive acute lymphoblastic leukemia and potentially defines subgroups with distinct clinical characteristics. (Cancer Sci 2008; 99: 1865–1868)


Nature Communications | 2016

Homeobox NKX2-3 promotes marginal-zone lymphomagenesis by activating B-cell receptor signalling and shaping lymphocyte dynamics

Eloy F. Robles; Maria Mena-Varas; Laura Barrio; Sara V. Merino-Cortes; Péter Balogh; Ming-Qing Du; Takashi Akasaka; Anton Parker; Sergio Roa; Carlos Panizo; Idoia Martin-Guerrero; Reiner Siebert; Victor Segura; Xabier Agirre; Laura Macri-Pellizeri; Beatriz Aldaz; Amaia Vilas-Zornoza; Shaowei Zhang; Sarah Moody; María José Calasanz; Thomas Tousseyn; Cyril Broccardo; Pierre Brousset; Elena Campos-Sanchez; Cesar Cobaleda; Isidro Sánchez-García; Jose Luis Fernandez-Luna; Ricardo García-Muñoz; Esther Pena; Beatriz Bellosillo

NKX2 homeobox family proteins have a role in cancer development. Here we show that NKX2-3 is overexpressed in tumour cells from a subset of patients with marginal-zone lymphomas, but not with other B-cell malignancies. While Nkx2-3-deficient mice exhibit the absence of marginal-zone B cells, transgenic mice with expression of NKX2-3 in B cells show marginal-zone expansion that leads to the development of tumours, faithfully recapitulating the principal clinical and biological features of human marginal-zone lymphomas. NKX2-3 induces B-cell receptor signalling by phosphorylating Lyn/Syk kinases, which in turn activate multiple integrins (LFA-1, VLA-4), adhesion molecules (ICAM-1, MadCAM-1) and the chemokine receptor CXCR4. These molecules enhance migration, polarization and homing of B cells to splenic and extranodal tissues, eventually driving malignant transformation through triggering NF-κB and PI3K-AKT pathways. This study implicates oncogenic NKX2-3 in lymphomagenesis, and provides a valid experimental mouse model for studying the biology and therapy of human marginal-zone B-cell lymphomas.


Nature Communications | 2017

Discovery of first-in-class reversible dual small molecule inhibitors against G9a and DNMTs in hematological malignancies.

Edurne San José-Enériz; Xabier Agirre; Obdulia Rabal; Amaia Vilas-Zornoza; Juan A. Sánchez-Arias; Estíbaliz Miranda; Ana Ugarte; Sergio Roa; Bruno Paiva; Ander Estella-Hermoso de Mendoza; Rosa Alvarez; Noelia Casares; Victor Segura; José I. Martín-Subero; François-Xavier Ogi; Pierre Soule; Clara M. Santiveri; Ramón Campos-Olivas; Giancarlo Castellano; Maite Garcia Fernandez de Barrena; Juan R. Rodriguez-Madoz; María José García-Barchino; Juan José Lasarte; Matías A. Avila; Jose A. Martinez-Climent; Julen Oyarzabal; Felipe Prosper

The indisputable role of epigenetics in cancer and the fact that epigenetic alterations can be reversed have favoured development of epigenetic drugs. In this study, we design and synthesize potent novel, selective and reversible chemical probes that simultaneously inhibit the G9a and DNMTs methyltransferase activity. In vitro treatment of haematological neoplasia (acute myeloid leukaemia-AML, acute lymphoblastic leukaemia-ALL and diffuse large B-cell lymphoma-DLBCL) with the lead compound CM-272, inhibits cell proliferation and promotes apoptosis, inducing interferon-stimulated genes and immunogenic cell death. CM-272 significantly prolongs survival of AML, ALL and DLBCL xenogeneic models. Our results represent the discovery of first-in-class dual inhibitors of G9a/DNMTs and establish this chemical series as a promising therapeutic tool for unmet needs in haematological tumours.


Journal of Medicinal Chemistry | 2018

Discovery of Reversible DNA Methyltransferase and Lysine Methyltransferase G9a Inhibitors with Antitumoral in Vivo Efficacy

Obdulia Rabal; Edurne San José-Enériz; Xabier Agirre; Juan A. Sánchez-Arias; Amaia Vilas-Zornoza; Ana Ugarte; Irene de Miguel; Estíbaliz Miranda; Leire Garate; Mario F. Fraga; Pablo Santamarina; Raúl F Pérez; Raquel Ordoñez; Elena Sáez; Sergio Roa; María José García-Barchino; Jose A. Martinez-Climent; Yingying Liu; Wei Wu; Musheng Xu; Felipe Prosper; Julen Oyarzabal

Using knowledge- and structure-based approaches, we designed and synthesized reversible chemical probes that simultaneously inhibit the activity of two epigenetic targets, histone 3 lysine 9 methyltransferase (G9a) and DNA methyltransferases (DNMT), at nanomolar ranges. Enzymatic competition assays confirmed our design strategy: substrate competitive inhibitors. Next, an initial exploration around our hit 11 was pursued to identify an adequate tool compound for in vivo testing. In vitro treatment of different hematological neoplasia cell lines led to the identification of molecules with clear antiproliferative efficacies (GI50 values in the nanomolar range). On the basis of epigenetic functional cellular responses (levels of lysine 9 methylation and 5-methylcytosine), an acceptable therapeutic window (around 1 log unit) and a suitable pharmacokinetic profile, 12 was selected for in vivo proof-of-concept ( Nat. Commun. 2017 , 8 , 15424 ). Herein, 12 achieved a significant in vivo efficacy: 70% overall tumor growth inhibition of a human acute myeloid leukemia (AML) xenograft in a mouse model.

Collaboration


Dive into the Amaia Vilas-Zornoza's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge