Network


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

Hotspot


Dive into the research topics where F. Javier Carmona is active.

Publication


Featured researches published by F. Javier Carmona.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Distinct DNA methylomes of newborns and centenarians

Holger Heyn; Ning Li; Humberto J. Ferreira; Sebastian Moran; David G. Pisano; Antonio Gomez; Javier Díez; Jose V. Sanchez-Mut; Fernando Setien; F. Javier Carmona; Annibale Alessandro Puca; Sergi Sayols; Miguel Angel Pujana; Jordi Serra-Musach; Isabel Iglesias-Platas; Francesc Formiga; Agustín F. Fernández; Mario F. Fraga; Simon Heath; Alfonso Valencia; Ivo Gut; Jun Wang; Manel Esteller

Human aging cannot be fully understood in terms of the constrained genetic setting. Epigenetic drift is an alternative means of explaining age-associated alterations. To address this issue, we performed whole-genome bisulfite sequencing (WGBS) of newborn and centenarian genomes. The centenarian DNA had a lower DNA methylation content and a reduced correlation in the methylation status of neighboring cytosine—phosphate—guanine (CpGs) throughout the genome in comparison with the more homogeneously methylated newborn DNA. The more hypomethylated CpGs observed in the centenarian DNA compared with the neonate covered all genomic compartments, such as promoters, exonic, intronic, and intergenic regions. For regulatory regions, the most hypomethylated sequences in the centenarian DNA were present mainly at CpG-poor promoters and in tissue-specific genes, whereas a greater level of DNA methylation was observed in CpG island promoters. We extended the study to a larger cohort of newborn and nonagenarian samples using a 450,000 CpG-site DNA methylation microarray that reinforced the observation of more hypomethylated DNA sequences in the advanced age group. WGBS and 450,000 analyses of middle-age individuals demonstrated DNA methylomes in the crossroad between the newborn and the nonagenarian/centenarian groups. Our study constitutes a unique DNA methylation analysis of the extreme points of human life at a single-nucleotide resolution level.


Journal of Clinical Oncology | 2013

A Prognostic DNA Methylation Signature for Stage I Non–Small-Cell Lung Cancer

Juan Sandoval; Jesús Méndez-González; Ernest Nadal; Guoan Chen; F. Javier Carmona; Sergi Sayols; Sebastian Moran; Holger Heyn; Miguel Vizoso; Antonio Gomez; Montse Sanchez-Cespedes; Yassen Assenov; Fabian Müller; Christoph Bock; Miquel Taron; Josefina Mora; Lucia Anna Muscarella; Triantafillos Liloglou; Michael P.A. Davies; Marina Pollán; Maria J. Pajares; Wenceslao Torre; Luis M. Montuenga; Elisabeth Brambilla; John K. Field; Luca Roz; Marco Lo Iacono; Giorgio V. Scagliotti; Rafael Rosell; David G. Beer

PURPOSE Non-small-cell lung cancer (NSCLC) is a tumor in which only small improvements in clinical outcome have been achieved. The issue is critical for stage I patients for whom there are no available biomarkers that indicate which high-risk patients should receive adjuvant chemotherapy. We aimed to find DNA methylation markers that could be helpful in this regard. PATIENTS AND METHODS A DNA methylation microarray that analyzes 450,000 CpG sites was used to study tumoral DNA obtained from 444 patients with NSCLC that included 237 stage I tumors. The prognostic DNA methylation markers were validated by a single-methylation pyrosequencing assay in an independent cohort of 143 patients with stage I NSCLC. RESULTS Unsupervised clustering of the 10,000 most variable DNA methylation sites in the discovery cohort identified patients with high-risk stage I NSCLC who had shorter relapse-free survival (RFS; hazard ratio [HR], 2.35; 95% CI, 1.29 to 4.28; P = .004). The study in the validation cohort of the significant methylated sites from the discovery cohort found that hypermethylation of five genes was significantly associated with shorter RFS in stage I NSCLC: HIST1H4F, PCDHGB6, NPBWR1, ALX1, and HOXA9. A signature based on the number of hypermethylated events distinguished patients with high- and low-risk stage I NSCLC (HR, 3.24; 95% CI, 1.61 to 6.54; P = .001). CONCLUSION The DNA methylation signature of NSCLC affects the outcome of stage I patients, and it can be practically determined by user-friendly polymerase chain reaction assays. The analysis of the best DNA methylation biomarkers improved prognostic accuracy beyond standard staging.


Carcinogenesis | 2013

DNA methylation profiling in breast cancer discordant identical twins identifies DOK7 as novel epigenetic biomarker

Holger Heyn; F. Javier Carmona; Antonio Gomez; Humberto J. Ferreira; Jordana T. Bell; Sergi Sayols; Kirsten Ward; Olafur A. Stefansson; Sebastian Moran; Juan Sandoval; Jorunn E. Eyfjörd; Tim D. Spector; Manel Esteller

Using whole blood from 15 twin pairs discordant for breast cancer and high-resolution (450K) DNA methylation analysis, we identified 403 differentially methylated CpG sites including known and novel potential breast cancer genes. Confirming the results in an independent validation cohort of 21 twin pairs determined the docking protein DOK7 as a candidate for blood-based cancer diagnosis. DNA hypermethylation of the promoter region was also seen in primary breast cancer tissues and cancer cell lines. Hypermethylation of DOK7 occurs years before tumor diagnosis, suggesting a role as a powerful epigenetic blood-based biomarker as well as providing insights into breast cancer pathogenesis.


Circulation-cardiovascular Genetics | 2014

A DNA Methylation Map of Human Atherosclerosis.

Silvio Zaina; Holger Heyn; F. Javier Carmona; Nuray Varol; Sergi Sayols; Enric Condom; José Ramírez-Ruz; Antonio Gomez; Isabel Gonçalves; Sebastian Moran; Manel Esteller

Background—Epigenetic alterations may contribute to the development of atherosclerosis. In particular, DNA methylation, a reversible and highly regulated DNA modification, could influence disease onset and progression because it functions as an effector for environmental influences, including diet and lifestyle, both of which are risk factors for cardiovascular diseases. Methods and Results—To address the role of DNA methylation changes in atherosclerosis, we compared a donor-matched healthy and atherosclerotic human aorta sample using whole-genome shotgun bisulfite sequencing. We observed that the atherosclerotic portion of the aorta was hypermethylated across many genomic loci in comparison with the matched healthy counterpart. Furthermore, we defined specific loci of differential DNA methylation using a set of donor-matched aortic samples and a high-density (>450 000 CpG sites) DNA methylation microarray. The functional importance in the disease was corroborated by crossing the DNA methylation signature with the corresponding expression data of the same samples. Among the differentially methylated CpGs associated with atherosclerosis onset, we identified genes participating in endothelial and smooth muscle functions. These findings provide new clues toward a better understanding of the molecular mechanisms of atherosclerosis. Conclusions—Our data identify an atherosclerosis-specific DNA methylation profile that highlights the contribution of different genes and pathways to the disorder. Interestingly, the observed gain of DNA methylation in the atherosclerotic lesions justifies efforts to develop DNA demethylating agents for therapeutic benefit.


Cancer Prevention Research | 2013

DNA Methylation Biomarkers for Noninvasive Diagnosis of Colorectal Cancer

F. Javier Carmona; Daniel Azuara; Antonio Berenguer-Llergo; Agustín F. Fernández; Sebastiano Biondo; Javier de Oca; Francisco Rodriguez-Moranta; Ramon Salazar; Alberto Villanueva; Mario F. Fraga; Jordi Guardiola; Gabriel Capellá; Manel Esteller; Victor Moreno

DNA methylation biomarkers for noninvasive diagnosis of colorectal cancer (CRC) and precursor lesions have been extensively studied. Different panels have been reported attempting to improve current protocols in clinical practice, although no definite biomarkers have been established. In the present study, we have examined patient biopsies starting from a comprehensive analysis of DNA methylation differences between paired normal and tumor samples in known cancer-related genes aiming to select the best performing candidates informative for CRC diagnosis in stool samples. Five selected markers were considered for subsequent analyses in independent biologic cohorts and in silico data sets. Among the five selected genes, three of them (AGTR1, WNT2 and SLIT2) were validated in stool DNA of affected patients with a detection sensitivity of 78% [95% confidence interval (CI), 56%–89%]. As a reference, DNA methylation of VIM and SEPT9 was evaluated in a subset of stool samples yielding sensitivities of 55% and 20%, respectively. Moreover, our panel may complement histologic and endoscopic diagnosis of inflammatory bowel disease (IBD)-associated neoplasia, as it was also efficient detecting aberrant DNA methylation in non-neoplastic tissue samples from affected patients. This novel panel of specific methylation markers can be useful for early diagnosis of CRC using stool DNA and may help in the follow-up of high-risk patients with IBD. Cancer Prev Res; 6(7); 656–65. ©2013 AACR.


Journal of the National Cancer Institute | 2014

Epigenetic Inactivation of the BRCA1 Interactor SRBC and Resistance to Oxaliplatin in Colorectal Cancer

Catia Moutinho; Anna Martínez-Cardús; Cristina Santos; Valentín Navarro-Perez; Eva Martinez-Balibrea; Eva Musulen; F. Javier Carmona; Andrea Sartore-Bianchi; Andrea Cassingena; Salvatore Siena; Elena Elez; Josep Tabernero; Ramon Salazar; Albert Abad; Manel Esteller

Background A major problem in cancer chemotherapy is the existence of primary resistance and/or the acquisition of secondary resistance. Many cellular defects contribute to chemoresistance, but epigenetic changes can also be a cause. Methods A DNA methylation microarray was used to identify epigenetic differences in oxaliplatin-sensitive and -resistant colorectal cancer cells. The candidate gene SRBC was validated by single-locus DNA methylation and expression techniques. Transfection and short hairpin experiments were used to assess oxaliplatin sensitivity. Progression-free survival (PFS) and overall survival (OS) in metastasic colorectal cancer patients were explored with Kaplan–Meier and Cox regression analyses. All statistical tests were two-sided. Results We found that oxaliplatin resistance in colorectal cancer cells depends on the DNA methylation–associated inactivation of the BRCA1 interactor SRBC gene. SRBC overexpression or depletion gives rise to sensitivity or resistance to oxaliplatin, respectively. SRBC epigenetic inactivation occurred in primary tumors from a discovery cohort of colorectal cancer patients (29.8%; n = 39 of 131), where it predicted shorter PFS (hazard ratio [HR] = 1.83; 95% confidence interval [CI] = 1.15 to 2.92; log-rank P = .01), particularly in oxaliplatin-treated case subjects for which metastasis surgery was not indicated (HR = 1.96; 95% CI = 1.13 to 3.40; log-rank P = .01). In a validation cohort of unresectable colorectal tumors treated with oxaliplatin (n = 58), SRBC hypermethylation was also associated with shorter PFS (HR = 1.90; 95% CI = 1.01 to 3.60; log-rank P = .045). Conclusions These results provide a basis for future clinical studies to validate SRBC hypermethylation as a predictive marker for oxaliplatin resistance in colorectal cancer.


Mutation Research | 2010

Epigenomics of human colon cancer.

F. Javier Carmona; Manel Esteller

DNA methylation exerts critical effects on gene-expression regulation in normal cells, and alterations on the normal methylation patterns characterize, and likely contribute, to cancer development and progression. The complete understanding of the distinctiveness, role and dynamics of the cancer cell epigenome has been hindered so far by technical limitations, and new, whole-genome approaches are required. Here we review the methods that have permitted us glimpses of the colon cancer methylome and outline future directions that will eventually lead researchers to identify the altered epigenetic patterns in cancer cells.


Cancer Cell | 2016

PDK1-SGK1 Signaling Sustains AKT-Independent mTORC1 Activation and Confers Resistance to PI3Kα Inhibition

Pau Castel; Haley Ellis; Ruzica Bago; Eneda Toska; Pedram Razavi; F. Javier Carmona; Srinivasaraghavan Kannan; Chandra Verma; Maura N. Dickler; Sarat Chandarlapaty; Edi Brogi; Dario R. Alessi; José Baselga; Maurizio Scaltriti

Summary PIK3CA, which encodes the p110α subunit of PI3K, is frequently mutated and oncogenic in breast cancer. PI3Kα inhibitors are in clinical development and despite promising early clinical activity, intrinsic resistance is frequent among patients. We have previously reported that residual downstream mTORC1 activity upon treatment with PI3Kα inhibitors drives resistance to these agents. However, the mechanism underlying this phenotype is not fully understood. Here we show that in cancer cells resistant to PI3Kα inhibition, PDK1 blockade restores sensitivity to these therapies. SGK1, which is activated by PDK1, contributes to the maintenance of residual mTORC1 activity through direct phosphorylation and inhibition of TSC2. Targeting either PDK1 or SGK1 prevents mTORC1 activation, restoring the antitumoral effects of PI3Kα inhibition in resistant cells.


Cancer Research | 2014

A Comprehensive DNA Methylation Profile of Epithelial-to-Mesenchymal Transition

F. Javier Carmona; Veronica Davalos; Enrique Vidal; Antonio Gomez; Holger Heyn; Yutaka Hashimoto; Miguel Vizoso; Anna Martínez-Cardús; Sergi Sayols; Humberto J. Ferreira; Jose V. Sanchez-Mut; Sebastian Moran; Mireia Margelí; Eva Castella; María Berdasco; Olafur A. Stefansson; Jorunn E. Eyfjörd; Eva González-Suárez; Joaquín Dopazo; Modesto Orozco; Ivo Gut; Manel Esteller

Epithelial-to-mesenchymal transition (EMT) is a plastic process in which fully differentiated epithelial cells are converted into poorly differentiated, migratory and invasive mesenchymal cells, and it has been related to the metastasis potential of tumors. This is a reversible process and cells can also eventually undergo mesenchymal-to-epithelial transition. The existence of a dynamic EMT process suggests the involvement of epigenetic shifts in the phenotype. Herein, we obtained the DNA methylomes at single-base resolution of Madin-Darby canine kidney cells undergoing EMT and translated the identified differentially methylated regions to human breast cancer cells undergoing a gain of migratory and invasive capabilities associated with the EMT phenotype. We noticed dynamic and reversible changes of DNA methylation, both on promoter sequences and gene-bodies in association with transcription regulation of EMT-related genes. Most importantly, the identified DNA methylation markers of EMT were present in primary mammary tumors in association with the epithelial or the mesenchymal phenotype of the studied breast cancer samples.


Science Translational Medicine | 2016

Somatic PIK3CA mutations as a driver of sporadic venous malformations

Pau Castel; F. Javier Carmona; Joaquim Grego-Bessa; Michael F. Berger; Agnes Viale; Kathryn V. Anderson; Silvia Bagué; Maurizio Scaltriti; Cristina R. Antonescu; Eulàlia Baselga; José Baselga

PIK3CA mutations induce venous malformations. PI3K-ing the best treatment Venous malformations are a type of congenital vascular anomalies composed of dilated blood vessels, which can cause a variety of complications such as pain, disfigurement, and bleeding. The available treatments for these malformations are invasive and not particularly effective. Now, Castel et al. and Castillo et al. have both identified mutations in the phosphatidylinositol 3-kinase (PI3K) pathway as a cause of venous malformations, studied these in numerous mouse models, and demonstrated that they can be effectively treated by inhibiting PI3K activity, paving the way for future clinical trials. Venous malformations (VM) are vascular malformations characterized by enlarged and distorted blood vessel channels. VM grow over time and cause substantial morbidity because of disfigurement, bleeding, and pain, representing a clinical challenge in the absence of effective treatments (Nguyen et al., 2014; Uebelhoer et al., 2012). Somatic mutations may act as drivers of these lesions, as suggested by the identification of TEK mutations in a proportion of VM (Limaye et al., 2009). We report that activating PIK3CA mutations gives rise to sporadic VM in mice, which closely resemble the histology of the human disease. Furthermore, we identified mutations in PIK3CA and related genes of the PI3K (phosphatidylinositol 3-kinase)/AKT pathway in about 30% of human VM that lack TEK alterations. PIK3CA mutations promote downstream signaling and proliferation in endothelial cells and impair normal vasculogenesis in embryonic development. We successfully treated VM in mouse models using pharmacological inhibitors of PI3Kα administered either systemically or topically. This study elucidates the etiology of a proportion of VM and proposes a therapeutic approach for this disease.

Collaboration


Dive into the F. Javier Carmona's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Holger Heyn

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar

Maurizio Scaltriti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Silvio Zaina

Universidad de Guanajuato

View shared research outputs
Top Co-Authors

Avatar

Anna Martínez-Cardús

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Eneda Toska

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge