Higinio Dopeso
Autonomous University of Barcelona
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Higinio Dopeso.
Cancer Research | 2006
Veronica Davalos; Higinio Dopeso; Julio Castaño; Andrew J. Wilson; Felip Vilardell; Jordi Romero-Giménez; Eloy Espín; Manel Armengol; Gabriel Capellá; John M. Mariadason; Lauri A. Aaltonen; Simó Schwartz; Diego Arango
The family of receptor tyrosine kinases EPH and their Ephrin ligands regulate cell proliferation, migration, and attachment. An important role in colorectal carcinogenesis is emerging for some of its members. In this study, we evaluate the role of EPHB4 in colorectal cancer and its value as a prognostic marker. EPHB4 levels were assessed by immunohistochemical staining of tissue microarrays of 137 colorectal tumors and aberrant hypermethylation of the EPHB4 promoter was investigated using methylation-specific PCR. We found that EPHB4 expression is frequently reduced or lost in colorectal tumors. Patients with low EPHB4 tumor levels had significantly shorter survival than patients in the high EPHB4 group (median survival, 1.8 and >9 years, respectively; P < 0.01, log-rank test), and this finding was validated using an independent set of 125 tumor samples. In addition, we show that EPHB4 promoter hypermethylation is a common mechanism of EPHB4 inactivation. Moreover, reintroduction of EPHB4 resulted in a significant reduction in the clonogenic potential of EPHB4-deficient cells, whereas abrogation of EPHB4 in cells with high levels of this receptor lead to a significant increase in clonogenicity. In summary, we identified EPHB4 as a useful prognostic marker for colorectal cancer. In addition, we provide mechanistic evidence showing that promoter methylation regulates EPHB4 transcription and functional evidence that EPHB4 can regulate the long-term clonogenic potential of colorectal tumor cells, revealing EPHB4 as a potential new tumor suppressor gene in colorectal cancer.
Cancer Research | 2009
Higinio Dopeso; Silvia Mateo-Lozano; Rocco Mazzolini; Paulo Rodrigues; Laura Lagares-Tena; Julian Ceron; J Romero; Marielle Esteves; Stefania Landolfi; Javier Hernández-Losa; Julio Castaño; Andrew J. Wilson; Santiago Ramón y Cajal; John M. Mariadason; Simó Schwartz; Diego Arango
Colorectal cancer is the second cause of cancer-related death in the western world, and although the genetic and molecular mechanisms involved in the initiation and progression of these tumors are among the best characterized, there are significant gaps in our understanding of this disease. The role of EPHB signaling in colorectal cancer has only recently been realized. Here, we use animal models to investigate the role of EphB4 in intestinal tumorigenesis. Modulation of EPHB4 levels in colon cancer cell lines resulted in significant differences in tumor growth in a xenograft model, with low levels of EPHB4 associated with faster growth. In addition, using a genetic model of intestinal tumorigenesis where adenomatous polyposis coli (Apc) mutations lead to initiation of the tumorigenic process (Apc(min) mice), we show that inactivation of a single allele of EphB4 results in higher proliferation in both the normal epithelium and intestinal tumors, significantly larger tumors in the small intestine, and a 10-fold increase in the number of tumors in the large intestine. This was associated with a 25% reduction in the lifespan of Apc(min) mice (P < 0.0001). Gene expression analysis showed that EphB4 mutations result in a profound transcriptional reprogramming, affecting genes involved in cell proliferation, remodeling of the extracellular matrix, and cell attachment to the basement membrane among other functional groups of genes. Importantly, in agreement with the expression profiling experiments, using an in vitro assay, we show that loss of EPHB4 in colon cancer cells results in a significantly increased potential to invade through a complex extracellular matrix. Collectively, these results indicate that EphB4 has tumor suppressor activities and that regulation of cell proliferation, extracellular matrix remodeling, and invasive potential are important mechanisms of tumor suppression.
Nature Communications | 2014
Paulo Rodrigues; Irati Macaya; Sarah Bazzocco; Rocco Mazzolini; Elena Andretta; Higinio Dopeso; Silvia Mateo-Lozano; Josipa Bilic; Fernando Cartón-García; Rocio Nieto; Lucia Suárez-López; Elsa Afonso; Stefania Landolfi; Javier Hernández-Losa; Kazuto Kobayashi; Santiago Ramón y Cajal; Josep Tabernero; Niall C. Tebbutt; John M. Mariadason; Simó Schwartz; Diego Arango
Activation of the small GTPase RHOA has strong oncogenic effects in many tumor types, although its role in colorectal cancer remains unclear. Here we show that RHOA inactivation contributes to colorectal cancer progression/metastasis, largely through the activation of Wnt/β-catenin signaling. RhoA inactivation in the murine intestine accelerates the tumorigenic process and in human colon cancer cells leads to the redistribution of β-catenin from the membrane to the nucleus and enhanced Wnt/β-catenin signaling, resulting in increased proliferation, invasion and de-differentiation. In mice, RHOA inactivation contributes to colon cancer metastasis and reduced RHOA levels were observed at metastatic sites compared to primary human colon tumors. Therefore, we have identified a new mechanism of activation of Wnt/β-catenin signaling and characterized the role of RHOA as a novel tumor suppressor in colorectal cancer. These results constitute a shift from the current paradigm and demonstrate that RHO GTPases can suppress tumor progression and metastasis.
Oncogene | 2007
Veronica Davalos; Higinio Dopeso; Sérgia Velho; Ana Ferreira; Luis Cirnes; N. Diaz-Chico; C. Bilbao; R. Ramirez; G. Rodriguez; O. Falcon; L. Leon; Renée C. Niessen; Gisela Keller; G. Dallenbach-Hellweg; Eloi Espín; Manel Armengol; A. Plaja; M. Perucho; Kohzoh Imai; Hiroyuki Yamamoto; Johannes Gebert; J. C. Diaz-Chico; Robert M. W. Hofstra; Stefan M. Woerner; Raquel Seruca; Simó Schwartz; Diego Arango
The EPH/EFN family of receptor tyrosine kinases regulates cell adhesion and migration and has an important role in controlling cell positioning in the normal intestinal epithelium. Inactivation of EPHB2 has recently been shown to accelerate tumorigenesis in the colon and rectum, and we have previously demonstrated frequent frameshift mutations (41%) in an A9 coding microsatellite repeat in exon 17 of EPHB2 in colorectal tumors with microsatellite instability (MSI). In this study, we extended these analyses to extracolonic MSI cancers, and found frameshift EPHB2 mutations in 39% (25/64) of gastric tumors and 14% (8/56) of endometrial tumors. Regression analysis of these EPHB2 mutation data on the basis of our previously proposed statistical model identified EPHB2 as a selective target of frameshift mutations in MSI gastric cancers but not in MSI endometrial carcinomas. These results suggest a functional role for EPHB2 in gastric tumor progression, and emphasize the differences between the tumorigenic processes in MSI gastrointestinal and endometrial cancer.
Clinical Cancer Research | 2010
Higinio Dopeso; Silvia Mateo-Lozano; Elena Elez; Stefania Landolfi; Francisco Javier Ramos Pascual; Javier Hernández-Losa; Rocco Mazzolini; Paulo Rodrigues; Sarah Bazzocco; Maria Josep Carreras; Eloy Espín; Manel Armengol; Andrew J. Wilson; John M. Mariadason; Santiago Ramón y Cajal; Josep Tabernero; Simó Schwartz; Diego Arango
Purpose: Irinotecan (CPT11) treatment significantly improves the survival of colorectal cancer patients and is routinely used for the treatment of these patients, alone or in combination with other agents. However, only 20% to 30% of patients show an objective response to irinotecan, and there is great need for new molecular markers capable of identifying the subset of patients who are unlikely to respond. Experimental Design: Here we used microarray analysis of a panel of 30 colorectal cancer cell lines and immunohistochemistry to identify and validate a new biomarker of response to irinotecan. Results: A good correlation was observed between irinotecan sensitivity and the expression of aprataxin (APTX), a histidine triad domain superfamily protein involved in DNA repair. Moreover, using an isogenic in vitro system deficient in APTX, we show that aprataxin directly regulates the cellular sensitivity to camptothecin, suggesting that it could be used to predict patient response to irinotecan. We constructed a tissue microarray containing duplicate tumor samples from 135 patients that received irinotecan for the treatment of advanced colorectal cancer. Immunohistochemical assessment of the tumor levels of aprataxin showed a significant association with treatment response and patient survival. Patients with low aprataxin had longer progression-free (9.2 versus 5.5 months; P = 0.03) and overall survival (36.7 versus 19.0 months; P = 0.008) than patients with high tumor aprataxin. No associations were found between coding APTX variants and aprataxin levels or camptothecin sensitivity. Conclusions: These results show that aprataxin tumor levels can be used to identify patients with low probability of response to irinotecan-based therapy who are ideal candidates to receive treatment with alternative agents in an attempt to improve patient survival. Clin Cancer Res; 16(8); 2375–82. ©2010 AACR.
Proceedings of the National Academy of Sciences of the United States of America | 2012
Rocco Mazzolini; Higinio Dopeso; Silvia Mateo-Lozano; Wakam Chang; Paulo Rodrigues; Sarah Bazzocco; Hafid Alazzouzi; Stefania Landolfi; Javier Hernández-Losa; Elena Andretta; Pia Alhopuro; Eloy Espín; Manel Armengol; Josep Tabernero; Santiago Ramón y Cajal; Matthias Kloor; Johannes Gebert; John M. Mariadason; Simó Schwartz; Lauri A. Aaltonen; Mark S. Mooseker; Diego Arango
The loss of the epithelial architecture and cell polarity/differentiation is known to be important during the tumorigenic process. Here we demonstrate that the brush border protein Myosin Ia (MYO1A) is important for polarization and differentiation of colon cancer cells and is frequently inactivated in colorectal tumors by genetic and epigenetic mechanisms. MYO1A frame-shift mutations were observed in 32% (37 of 116) of the colorectal tumors with microsatellite instability analyzed, and evidence of promoter methylation was observed in a significant proportion of colon cancer cell lines and primary colorectal tumors. The loss of polarization/differentiation resulting from MYO1A inactivation is associated with higher tumor growth in soft agar and in a xenograft model. In addition, the progression of genetically and carcinogen-initiated intestinal tumors was significantly accelerated in Myo1a knockout mice compared with Myo1a wild-type animals. Moreover, MYO1A tumor expression was found to be an independent prognostic factor for colorectal cancer patients. Patients with low MYO1A tumor protein levels had significantly shorter disease-free and overall survival compared with patients with high tumoral MYO1A (logrank test P = 0.004 and P = 0.009, respectively). The median time-to-disease recurrence in patients with low MYO1A was 1 y, compared with >9 y in the group of patients with high MYO1A. These results identify MYO1A as a unique tumor-suppressor gene in colorectal cancer and demonstrate that the loss of structural brush border proteins involved in cell polarity are important for tumor development.
International Journal of Cancer | 2008
Jordi Romero-Giménez; Higinio Dopeso; Ignacio Blanco; Angel Guerra-Moreno; Sara González; Stefanie Vogt; Stefan Aretz; Simó Schwartz; Gabriel Capellá; Diego Arango
Familial adenomatous polyposis (FAP) is an autosomal dominant syndrome predisposing to colorectal cancer and affects 1 in 5–10,000 births. Inheritance of a mutant allele of the adenomatous polyposis coli (APC) gene is the cause of ∼80% of FAP and 20–30% of an attenuated form of FAP (AFAP), whereas mutations in MUTYH account for a small proportion of the remaining cases. However, the genetic cause of FAP/AFAP in a significant number of families is not known, and cancer risk for individual members of these families cannot be assessed. There is, therefore, an acute need to identify the underlying genetic cause responsible for FAP/AFAP in APC/MUTYH mutation negative families. Hypermethylation of CpG islands in the promoter of tumor suppressor genes can result in gene silencing, has been shown to be functionally equivalent to genetic mutations and can be inherited. Moreover, APC promoter hypermethylation is observed in ∼20% of sporadic colorectal tumors and correlates with the loss of gene expression. In our study, we used bisulfite treatment and direct sequencing of 2 regulatory regions of APC containing a total of 25 CpG dinucleotides, to investigate the possible role of germline hypermethylation of the APC promoter in FAP and AFAP families that were negative for APC and MUTYH mutations. Analysis of 21 FAP and 39 AFAP families did not identify signs of abnormal promoter methylation, indicating that this form of epigenetic silencing is not a common cause of FAP/AFAP. These results substantially contribute to clarify the potential role of germline epimutations as a cause of inherited predisposition to cancer.
American Journal of Pathology | 2012
Diego Arango; Sheren Al-Obaidi; David S. Williams; Higinio Dopeso; Rocco Mazzolini; Georgia A. Corner; Do-Sun Byun; Azadeh Carr; Carmel Murone; Lars Tögel; Nikolajs Zeps; Lauri A. Aaltonen; Barry Iacopetta; John M. Mariadason
Colorectal cancers (CRCs) are classified as having microsatellite instability (MSI) or chromosomal instability (CIN); herein termed microsatellite stable (MSS). MSI colon cancers frequently display a poorly differentiated histology for which the molecular basis is not well understood. Gene expression and immunohistochemical profiling of MSS and MSI CRC cell lines and tumors revealed significant down-regulation of the intestinal-specific cytoskeletal protein villin in MSI colon cancer, with complete absence in 62% and 17% of MSI cell lines and tumors, respectively. Investigation of 577 CRCs linked loss of villin expression to poorly differentiated histology in MSI and MSS tumors. Furthermore, mislocalization of villin from the membrane was prognostic for poorer outcome in MSS patients. Loss of villin expression was not due to coding sequence mutations, epigenetic inactivation, or promoter mutation. Conversely, in transient transfection assays villin promoter activity reflected endogenous villin expression, suggesting transcriptional control. A screen of gut-specific transcription factors revealed a significant correlation between expression of villin and the homeobox transcription factor Cdx-1. Cdx-1 overexpression induced villin promoter activity, Cdx-1 knockdown down-regulated endogenous villin expression, and deletion of a key Cdx-binding site within the villin promoter attenuated promoter activity. Loss of Cdx-1 expression in CRC lines was associated with Cdx-1 promoter methylation. These findings demonstrate that loss of villin expression due to Cdx-1 loss is a feature of poorly differentiated CRCs.
International Journal of Cancer | 2013
Rocco Mazzolini; Paulo Rodrigues; Sarah Bazzocco; Higinio Dopeso; Ana Ferreira; Silvia Mateo-Lozano; Elena Andretta; Stefan M. Woerner; Hafid Alazzouzi; Stefania Landolfi; Javier Hernández-Losa; Irati Macaya; Hiromu Suzuki; Santiago Ramón y Cajal; Mark S. Mooseker; John M. Mariadason; Johannes Gebert; Robert M.W. Hofstra; Jaume Reventós; Hiroyuki Yamamoto; Simó Schwartz; Diego Arango
Brush border Myosin Ia (MYO1A) has been shown to be frequently mutated in colorectal tumors with microsatellite instability (MSI) and to have tumor suppressor activity in intestinal tumors. Here, we investigated the frequency of frameshift mutations in the A8 microsatellite in exon 28 of MYO1A in MSI gastric and endometrial tumors and found a high mutation rate in gastric (22/47; 46.8%) but not endometrial (3/48; 6.2%) tumors. Using a regression model, we show that MYO1A mutations are likely to confer a growth advantage to gastric, but not endometrial tumors. The mutant MYO1A7A protein was shown to lose its membrane localization in gastric cancer cells and a cycloheximide‐chase assay demonstrated that the mutant MYO1A7A protein has reduced stability compared to the wild type MYO1A. Frequent MYO1A promoter hypermethylation was also found in gastric tumors. Promoter methylation negatively correlates with MYO1A mRNA expression in a series of 58 non‐MSI gastric primary tumors (Pearsons r = −0.46; p = 0.0003) but not in a cohort of 54 non‐MSI endometrial tumors and treatment of gastric cancer cells showing high MYO1A promoter methylation with the demethylating agent 5‐aza‐2′‐deoxycytidine, resulted in a significant increase of MYO1A mRNA levels. We found that normal gastric epithelial cells, but not normal endometrial cells, express high levels of MYO1A. Therefore, when considered together, our findings suggest that MYO1A has tumor suppressor activity in the normal gastric epithelium but not in the normal endometrium and inactivation of MYO1A either genetically or epigenetically may confer gastric epithelial cells a growth advantage.
Scientific Reports | 2017
Silvia Mateo-Lozano; Sarah Bazzocco; Paulo Rodrigues; Rocco Mazzolini; Elena Andretta; Higinio Dopeso; Yolanda Fernández; Edgar del Llano; Josipa Bilic; Lucia Suárez-López; Irati Macaya; Fernando Cartón-García; Rocio Nieto; Lizbeth M. Jimenez-Flores; Priscila Guimarães de Marcondes; Yaiza Nuñez; Elsa Afonso; Karina Cacci; Javier Hernández-Losa; Stefania Landolfi; Ibane Abasolo; Santiago Ramón y Cajal; John M. Mariadason; Simó Schwartz; Toshimitsu Matsui; Diego Arango
Although deregulation of EPHB signaling has been shown to be an important step in colorectal tumorigenesis, the role of EPHB6 in this process has not been investigated. We found here that manipulation of EPHB6 levels in colon cancer cell lines has no effect on their motility and growth on a solid substrate, soft agar or in a xenograft mouse model. We then used an EphB6 knockout mouse model to show that EphB6 inactivation does not efficiently initiate tumorigenesis in the intestinal tract. In addition, when intestinal tumors are initiated genetically or pharmacologically in EphB6+/+ and EphB6−/− mice, no differences were observed in animal survival, tumor multiplicity, size or histology, and proliferation of intestinal epithelial cells or tumor cells. However, reintroduction of EPHB6 into colon cancer cells significantly reduced the number of lung metastasis after tail-vein injection in immunodeficient mice, while EPHB6 knockdown in EPHB6-expressing cells increased their metastatic spread. Consistently, although EPHB6 protein expression in a series of 130 primary colorectal tumors was not associated with patient survival, EPHB6 expression was significantly lower in lymph node metastases compared to primary tumors. Our results indicate that the loss of EPHB6 contributes to the metastatic process of colorectal cancer.