11 results on '"San José-Eneriz E"'
Search Results
2. Epigenetic Regulation of MicroRNAs in Acute Lymphoblastic Leukemia.
- Author
-
Roman-Gomez J, Agirre X, Jiménez-Velasco A, Arqueros V, Vilas-Zornoza A, Rodriguez-Otero P, Martin-Subero I, Garate L, Cordeu L, San José-Eneriz E, Martin V, Castillejo JA, Bandrés E, Calasanz MJ, Siebert R, Heiniger A, Torres A, and Prosper F
- Published
- 2009
- Full Text
- View/download PDF
3. MicroRNA expression profiling in Imatinib-resistant Chronic Myeloid Leukemia patients without clinically significant ABL1-mutations
- Author
-
José Calasanz María, Prósper Felipe, Rodríguez-Otero Paula, Vilas-Zornoza Amaia, Cordeu Lucia, Martin Vanesa, Garate Leire, Jiménez-Velasco Antonio, Román-Gómez José, San José-Enériz Edurne, and Agirre Xabier
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract 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.
- Published
- 2009
- Full Text
- View/download PDF
4. Immunogenomic identification and characterization of granulocytic myeloid-derived suppressor cells in multiple myeloma.
- Author
-
Perez C, Botta C, Zabaleta A, Puig N, Cedena MT, Goicoechea I, Alameda D, San José-Eneriz E, Merino J, Rodríguez-Otero P, Maia C, Alignani D, Maiso P, Manrique I, Lara-Astiaso D, Vilas-Zornoza A, Sarvide S, Riillo C, Rossi M, Rosiñol L, Oriol A, Blanchard MJ, Rios R, Sureda A, Martin J, Martinez R, Bargay J, de la Rubia J, Hernandez MT, Martinez-Lopez J, Orfao A, Agirre X, Prosper F, Mateos MV, Lahuerta JJ, Blade J, San-Miguel JF, and Paiva B
- Subjects
- Female, Follow-Up Studies, Humans, Lymphocyte Count, Male, Middle Aged, Neutrophils immunology, Neutrophils metabolism, Neutrophils pathology, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes pathology, Transcription, Genetic immunology, Antigens, CD blood, Antigens, CD genetics, Antigens, CD immunology, Multiple Myeloma blood, Multiple Myeloma genetics, Multiple Myeloma immunology, Multiple Myeloma pathology, Myeloid-Derived Suppressor Cells immunology, Myeloid-Derived Suppressor Cells metabolism, Myeloid-Derived Suppressor Cells pathology, Neoplasm Proteins blood, Neoplasm Proteins genetics, Neoplasm Proteins immunology
- Abstract
Granulocytic myeloid-derived suppressor cells (G-MDSCs) promote tumor growth and immunosuppression in multiple myeloma (MM). However, their phenotype is not well established for accurate monitoring or clinical translation. We aimed to provide the phenotypic profile of G-MDSCs based on their prognostic significance in MM, immunosuppressive potential, and molecular program. The preestablished phenotype of G-MDSCs was evaluated in bone marrow samples from controls and MM patients using multidimensional flow cytometry; surprisingly, we found that CD11b+CD14-CD15+CD33+HLADR- cells overlapped with common eosinophils and neutrophils, which were not expanded in MM patients. Therefore, we relied on automated clustering to unbiasedly identify all granulocytic subsets in the tumor microenvironment: basophils, eosinophils, and immature, intermediate, and mature neutrophils. In a series of 267 newly diagnosed MM patients (GEM2012MENOS65 trial), only the frequency of mature neutrophils at diagnosis was significantly associated with patient outcome, and a high mature neutrophil/T-cell ratio resulted in inferior progression-free survival (P < .001). Upon fluorescence-activated cell sorting of each neutrophil subset, T-cell proliferation decreased in the presence of mature neutrophils (0.5-fold; P = .016), and the cytotoxic potential of T cells engaged by a BCMA×CD3-bispecific antibody increased notably with the depletion of mature neutrophils (fourfold; P = .0007). Most interestingly, RNA sequencing of the 3 subsets revealed that G-MDSC-related genes were specifically upregulated in mature neutrophils from MM patients vs controls because of differential chromatin accessibility. Taken together, our results establish a correlation between the clinical significance, immunosuppressive potential, and transcriptional network of well-defined neutrophil subsets, providing for the first time a set of optimal markers (CD11b/CD13/CD16) for accurate monitoring of G-MDSCs in MM., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
5. Dual Targeting of Histone Methyltransferase G9a and DNA-Methyltransferase 1 for the Treatment of Experimental Hepatocellular Carcinoma.
- Author
-
Bárcena-Varela M, Caruso S, Llerena S, Álvarez-Sola G, Uriarte I, Latasa MU, Urtasun R, Rebouissou S, Alvarez L, Jimenez M, Santamaría E, Rodriguez-Ortigosa C, Mazza G, Rombouts K, San José-Eneriz E, Rabal O, Agirre X, Iraburu M, Santos-Laso A, Banales JM, Zucman-Rossi J, Prósper F, Oyarzabal J, Berasain C, Ávila MA, and Fernández-Barrena MG
- Subjects
- Animals, Antineoplastic Agents pharmacology, CCAAT-Enhancer-Binding Proteins metabolism, Carcinoma, Hepatocellular enzymology, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, Dogs, Hep G2 Cells, Histone-Lysine N-Methyltransferase metabolism, Humans, Liver Neoplasms, Experimental enzymology, Madin Darby Canine Kidney Cells, Male, Mice, Nude, Ubiquitin-Protein Ligases metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Carcinoma, Hepatocellular drug therapy, DNA (Cytosine-5-)-Methyltransferase 1 antagonists & inhibitors, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Liver Neoplasms, Experimental drug therapy
- Abstract
Epigenetic modifications such as DNA and histone methylation functionally cooperate in fostering tumor growth, including that of hepatocellular carcinoma (HCC). Pharmacological targeting of these mechanisms may open new therapeutic avenues. We aimed to determine the therapeutic efficacy and potential mechanism of action of our dual G9a histone-methyltransferase and DNA-methyltransferase 1 (DNMT1) inhibitor in human HCC cells and their crosstalk with fibrogenic cells. The expression of G9a and DNMT1, along with that of their molecular adaptor ubiquitin-like with PHD and RING finger domains-1 (UHRF1), was measured in human HCCs (n = 268), peritumoral tissues (n = 154), and HCC cell lines (n = 32). We evaluated the effect of individual and combined inhibition of G9a and DNMT1 on HCC cell growth by pharmacological and genetic approaches. The activity of our lead compound, CM-272, was examined in HCC cells under normoxia and hypoxia, human hepatic stellate cells and LX2 cells, and xenograft tumors formed by HCC or combined HCC+LX2 cells. We found a significant and correlative overexpression of G9a, DNMT1, and UHRF1 in HCCs in association with poor prognosis. Independent G9a and DNMT1 pharmacological targeting synergistically inhibited HCC cell growth. CM-272 potently reduced HCC and LX2 cells proliferation and quelled tumor growth, particularly in HCC+LX2 xenografts. Mechanistically, CM-272 inhibited the metabolic adaptation of HCC cells to hypoxia and induced a differentiated phenotype in HCC and fibrogenic cells. The expression of the metabolic tumor suppressor gene fructose-1,6-bisphosphatase (FBP1), epigenetically repressed in HCC, was restored by CM-272. Conclusion: Combined targeting of G9a/DNMT1 with compounds such as CM-272 is a promising strategy for HCC treatment. Our findings also underscore the potential of differentiation therapy in HCC., (© 2018 by the American Association for the Study of Liver Diseases.)
- Published
- 2019
- Full Text
- View/download PDF
6. In-silico gene essentiality analysis of polyamine biosynthesis reveals APRT as a potential target in cancer.
- Author
-
Pey J, San José-Eneriz E, Ochoa MC, Apaolaza I, de Atauri P, Rubio A, Cendoya X, Miranda E, Garate L, Cascante M, Carracedo A, Agirre X, Prosper F, and Planes FJ
- Subjects
- Adenosylmethionine Decarboxylase metabolism, Biochemical Phenomena, Cell Line, Tumor, Cell Proliferation, Computer Simulation, Genes, Essential genetics, Homeostasis, Humans, Leukemia genetics, Metabolic Networks and Pathways, Neoplasms genetics, Polyelectrolytes, Adenine Phosphoribosyltransferase metabolism, Adenine Phosphoribosyltransferase physiology, Polyamines metabolism
- Abstract
Constraint-based modeling for genome-scale metabolic networks has emerged in the last years as a promising approach to elucidate drug targets in cancer. Beyond the canonical biosynthetic routes to produce biomass, it is of key importance to focus on metabolic routes that sustain the proliferative capacity through the regulation of other biological means in order to improve in-silico gene essentiality analyses. Polyamines are polycations with central roles in cancer cell proliferation, through the regulation of transcription and translation among other things, but are typically neglected in in silico cancer metabolic models. In this study, we analysed essential genes for the biosynthesis of polyamines. Our analysis corroborates the importance of previously known regulators of the pathway, such as Adenosylmethionine Decarboxylase 1 (AMD1) and uncovers novel enzymes predicted to be relevant for polyamine homeostasis. We focused on Adenine Phosphoribosyltransferase (APRT) and demonstrated the detrimental consequence of APRT gene silencing on different leukaemia cell lines. Our results highlight the importance of revisiting the metabolic models used for in-silico gene essentiality analyses in order to maximize the potential for drug target identification in cancer.
- Published
- 2017
- Full Text
- View/download PDF
7. An in-silico approach to predict and exploit synthetic lethality in cancer metabolism.
- Author
-
Apaolaza I, San José-Eneriz E, Tobalina L, Miranda E, Garate L, Agirre X, Prósper F, and Planes FJ
- Subjects
- Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Gene Silencing, Genes, Neoplasm, Humans, Computer Simulation, Neoplasms genetics, Neoplasms metabolism, Synthetic Lethal Mutations genetics
- Abstract
Synthetic lethality is a promising concept in cancer research, potentially opening new possibilities for the development of more effective and selective treatments. Here, we present a computational method to predict and exploit synthetic lethality in cancer metabolism. Our approach relies on the concept of genetic minimal cut sets and gene expression data, demonstrating a superior performance to previous approaches predicting metabolic vulnerabilities in cancer. Our genetic minimal cut set computational framework is applied to evaluate the lethality of ribonucleotide reductase catalytic subunit M1 (RRM1) inhibition in multiple myeloma. We present a computational and experimental study of the effect of RRM1 inhibition in four multiple myeloma cell lines. In addition, using publicly available genome-scale loss-of-function screens, a possible mechanism by which the inhibition of RRM1 is effective in cancer is established. Overall, our approach shows promising results and lays the foundation to build a novel family of algorithms to target metabolism in cancer.Exploiting synthetic lethality is a promising approach for cancer therapy. Here, the authors present an approach to identifying such interactions by finding genetic minimal cut sets (gMCSs) that block cancer proliferation, and apply it to study the lethality of RRM1 inhibition in multiple myeloma.
- Published
- 2017
- Full Text
- View/download PDF
8. Frequent and simultaneous epigenetic inactivation of TP53 pathway genes in acute lymphoblastic leukemia.
- Author
-
Vilas-Zornoza A, Agirre X, Martín-Palanco V, Martín-Subero JI, San José-Eneriz E, Garate L, Álvarez S, Miranda E, Rodríguez-Otero P, Rifón J, Torres A, Calasanz MJ, Cruz Cigudosa J, Román-Gómez J, and Prósper F
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Cell Line, Tumor, Child, Child, Preschool, Cohort Studies, Female, Gene Expression Regulation, Leukemic, Gene Frequency, Humans, Infant, Male, Middle Aged, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Prognosis, Retrospective Studies, Signal Transduction genetics, Young Adult, Epigenesis, Genetic physiology, Gene Silencing physiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Aberrant DNA methylation is one of the most frequent alterations in patients with Acute Lymphoblastic Leukemia (ALL). Using methylation bead arrays we analyzed the methylation status of 807 genes implicated in cancer in a group of ALL samples at diagnosis (n = 48). We found that 154 genes were methylated in more than 10% of ALL samples. Interestingly, the expression of 13 genes implicated in the TP53 pathway was downregulated by hypermethylation. Direct or indirect activation of TP53 pathway with 5-aza-2'-deoxycitidine, Curcumin or Nutlin-3 induced an increase in apoptosis of ALL cells. The results obtained with the initial group of 48 patients was validated retrospectively in a second cohort of 200 newly diagnosed ALL patients. Methylation of at least 1 of the 13 genes implicated in the TP53 pathway was observed in 78% of the patients, which significantly correlated with a higher relapse (p = 0.001) and mortality (p<0.001) rate being an independent prognostic factor for disease-free survival (DFS) (p = 0.006) and overall survival (OS) (p = 0.005) in the multivariate analysis. All these findings indicate that TP53 pathway is altered by epigenetic mechanisms in the majority of ALL patients and correlates with prognosis. Treatments with compounds that may reverse the epigenetic abnormalities or activate directly the p53 pathway represent a new therapeutic alternative for patients with ALL.
- Published
- 2011
- Full Text
- View/download PDF
9. Epigenetic down-regulation of BIM expression is associated with reduced optimal responses to imatinib treatment in chronic myeloid leukaemia.
- Author
-
San José-Eneriz E, Agirre X, Jiménez-Velasco A, Cordeu L, Martín V, Arqueros V, Gárate L, Fresquet V, Cervantes F, Martínez-Climent JA, Heiniger A, Torres A, Prósper F, and Roman-Gomez J
- Subjects
- Adult, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Apoptosis Regulatory Proteins genetics, Azacitidine analogs & derivatives, Azacitidine pharmacology, Bcl-2-Like Protein 11, Benzamides, Cell Proliferation drug effects, DNA Methylation, DNA Modification Methylases antagonists & inhibitors, Decitabine, Dose-Response Relationship, Drug, Down-Regulation drug effects, Drug Evaluation, Preclinical methods, Drug Resistance, Neoplasm genetics, Epigenesis, Genetic, Female, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic genetics, Humans, Imatinib Mesylate, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Male, Membrane Proteins genetics, Middle Aged, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins genetics, RNA, Messenger genetics, RNA, Neoplasm genetics, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Apoptosis Regulatory Proteins biosynthesis, Down-Regulation genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Membrane Proteins biosynthesis, Piperazines pharmacology, Proto-Oncogene Proteins biosynthesis, Pyrimidines pharmacology
- Abstract
Background: Expression of the pro-apoptotic BCL-2-interacting mediator (BIM) has recently been implicated in imatinib-induced apoptosis of BCR-ABL1(+) cells. However, the mechanisms involved in the regulation of BIM in CML and its role in the clinical setting have not been established., Design and Methods: We analysed the mRNA expression of BIM in 100 newly diagnosed patients with CML in chronic phase by Q-RT-PCR and the protein levels by Western blot analysis. Methylation status was analysed by bisulphite genomic sequencing and MSP. CML cell lines were treated with imatinib and 5-aza-2'-deoxycytidine, and were transfected with two different siRNAs against BIM and cell proliferation and apoptosis were analysed., Results: We demonstrated that down-regulation of BIM expression was present in 36% of the patients and was significantly associated with a lack of optimal response to imatinib as indicated by the decrease in cytogenetic and molecular responses at 6, 12 and 18 months in comparison with patients with normal BIM expression (p<0.05). Expression of BIM was mediated by promoter hypermethylation as demonstrated by restoration of BIM expression after treatment of CML cells with 5-aza-2'-deoxycytidine. Using CML cell lines with low and normal expression of BIM we further demonstrated that the expression of BIM is required for imatinib-induced CML apoptosis., Conclusion: Our data indicate that down-regulation of BIM is epigenetically controlled by methylation in a percentage of CML patients and has an unfavourable prognostic impact, and that the combination of imatinib with a de-methylating agent may result in improved responses in patients with decreased expression of BIM.
- Published
- 2009
- Full Text
- View/download PDF
10. Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia.
- Author
-
Agirre X, Vilas-Zornoza A, Jiménez-Velasco A, Martin-Subero JI, Cordeu L, Gárate L, San José-Eneriz E, Abizanda G, Rodríguez-Otero P, Fortes P, Rifón J, Bandrés E, Calasanz MJ, Martín V, Heiniger A, Torres A, Siebert R, Román-Gomez J, and Prósper F
- Subjects
- Animals, Cell Division, Cell Line, Tumor, Chromatin genetics, Chromatin physiology, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Disease Models, Animal, Disease-Free Survival, Down-Regulation, Female, Humans, Male, Mice, Mice, Knockout, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Prognosis, RNA, Messenger genetics, Survival Rate, Transcription, Genetic, Cyclin-Dependent Kinase 6 genetics, Epigenesis, Genetic genetics, Gene Silencing, MicroRNAs genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
11. Epigenetic regulation of Wnt-signaling pathway in acute lymphoblastic leukemia.
- Author
-
Román-Gómez J, Cordeu L, Agirre X, Jiménez-Velasco A, San José-Eneriz E, Garate L, Calasanz MJ, Heiniger A, Torres A, and Prosper F
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Antimetabolites, Antineoplastic administration & dosage, Azacitidine administration & dosage, Azacitidine analogs & derivatives, Cell Line, Tumor, Child, Child, Preschool, DNA Methylation drug effects, Decitabine, Disease-Free Survival, Female, Follow-Up Studies, Gene Expression Regulation, Leukemic drug effects, Gene Expression Regulation, Leukemic genetics, Humans, Infant, Male, Middle Aged, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Quercetin administration & dosage, Wnt Proteins antagonists & inhibitors, Wnt Proteins genetics, beta Catenin genetics, Epigenesis, Genetic drug effects, Neoplasm Proteins metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Signal Transduction drug effects, Signal Transduction genetics, Wnt Proteins metabolism, beta Catenin metabolism
- Abstract
Activation of the Wnt/beta-catenin signaling pathway is a hallmark of a number of solid tumors. We analyzed the regulation of the Wnt/beta-catenin pathway in acute lymphoblastic leukemia (ALL) and its role in the pathogenesis of the disease. We found that expression of the Wnt inhibitors sFRP1, sFRP2, sFRP4, sFRP5, WIF1, Dkk3, and Hdpr1 was down-regulated due to abnormal promoter methylation in ALL cell lines and samples from patients with ALL. Methylation of Wnt inhibitors was associated with activation of the Wnt-signaling pathway as demonstrated by the up-regulation of the Wnt target genes WNT16, FZ3, TCF1, LEF1, and cyclin D1 in cell lines and samples and the nuclear localization of beta-catenin in cell lines. Treatment of ALL cells with the Wnt inhibitor quercetin or with the demethylating agent 5-aza-2'-deoxycytidine induced an inactivation of the Wnt pathway and induced apoptosis of ALL cells. Finally, in a group of 261 patients with newly diagnosed ALL, abnormal methylation of Wnt inhibitors was associated with decreased 10-year disease-free survival (25% versus 66% respectively, P < .001) and overall survival (28% versus 61% respectively, P = .001). Our results indicate a role of abnormal Wnt signaling in ALL and establish a group of patients with a significantly worse prognosis (methylated group).
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.