Network


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

Hotspot


Dive into the research topics where Marta I. Sierra is active.

Publication


Featured researches published by Marta I. Sierra.


Clinical Cancer Research | 2004

Protein Kinase C θ Is Highly Expressed in Gastrointestinal Stromal Tumors But Not in Other Mesenchymal Neoplasias

Pilar Blay; Aurora Astudillo; J. M. Buesa; Elias Campo; Mar Abad; Juan García-García; Rosa Miquel; Vicente Marco; Marta I. Sierra; Raquel Losa; A. J. Lacave; Alejandro Braña; Milagros Balbín; José M. P. Freije

Purpose: Gastrointestinal stromal tumors (GIST) are a distinctive group of mesenchymal neoplasms of the gastrointestinal tract. The oncogene KIT has a central role in the pathogenesis of GIST, with c-kit receptor tyrosine kinase (KIT) protein expression being the gold standard in its diagnosis. The identification of GIST patients has become crucial, because the tyrosine kinase inhibitor Imatinib is effective in the treatment of this malignancy. However, a small set of GISTs remain unrecognized, because KIT protein expression is not always evident. The aim of this study was the identification of new markers for the differential diagnosis of GIST. Experimental Design: By analyzing publicly available data from transcriptional profiling of sarcomas, we found that protein kinase C θ (PKC-θ), a novel PKC isotype involved in T-cell activation, is highly and specifically expressed in GIST. PKC-θ expression in GIST was confirmed by reverse transcription-PCR and Western blot. PKC-θ was analyzed by immunohistochemistry in a panel of 26 GIST, 12 non-GIST soft-tissue sarcomas, and 35 tumors from other histologies. Results: We found that all of the GISTs expressed PKC-θ, whereas this protein was undetectable in other mesenchymal or epithelial tumors, including non-GIST KIT-positive tumors. PKC-θ immunoreactivity was also observed in interstitial cells of Cajal. Conclusions: Our results show that PKC-θ is easily detected by immunohistochemistry in GIST specimens and that it could be a sensitive and specific marker for the diagnosis of this malignancy.


Cancer | 2004

Phase I clinical trial of fixed-dose rate infusional gemcitabine and dacarbazine in the treatment of advanced soft tissue sarcoma, with assessment of gemcitabine triphosphate accumulation

J. Buesa; Raquel Losa; Aida Fernández; Marta I. Sierra; E. Esteban; Ángela Díaz; Antonio Lopez-Pousa; Joaquin Fra

In the current study, the authors set out to determine the dose‐limiting toxicity (DLT) and maximum tolerated dose (MTD) associated with a combination of gemcitabine and dacarbazine (DTIC) in patients with advanced soft tissue sarcoma (ASTS), to obtain preliminary information on the activity of this combination, and to explore possible pharmacodynamic interactions between gemcitabine and DTIC.


Current Genomics | 2015

Epigenetics of Aging.

Marta I. Sierra; Agustín F. Fernández; Mario F. Fraga

The best-known phenomenon exemplifying epigenetic drift (the alteration of epigenetic patterns during aging) is the gradual decrease of global DNA methylation. Aging cells, different tissue types, as well as a variety of human diseases possess their own distinct DNA methylation profiles, although the functional impact of these is not always clear. DNA methylation appears to be a dynamic tool of transcriptional regulation, with an extra layer of complexity due to the recent discovery of the conversion of 5-methylcytosine into 5-hydroxymethylcytosine. This age-related DNA demethylation is associated with changes in histone modification patterns and, furthermore, we now know that ncRNAs have evolved in eukaryotes as epigenetic regulators of gene expression. In this review, we will discuss current knowledge on how all these epigenetic phenomena are implicated in human aging, and their links with external, internal and stochastic factors which can affect human age-related diseases onset.


International Journal of Nanomedicine | 2016

The effect of exposure to nanoparticles and nanomaterials on the mammalian epigenome

Marta I. Sierra; Adolfo Valdés; Agustín F. Fernández; Ramón Torrecillas; Mario F. Fraga

Human exposure to nanomaterials and nanoparticles is increasing rapidly, but their effects on human health are still largely unknown. Epigenetic modifications are attracting ever more interest as possible underlying molecular mechanisms of gene–environment interactions, highlighting them as potential molecular targets following exposure to nanomaterials and nanoparticles. Interestingly, recent research has identified changes in DNA methylation, histone post-translational modifications, and noncoding RNAs in mammalian cells exposed to nanomaterials and nanoparticles. However, the challenge for the future will be to determine the molecular pathways driving these epigenetic alterations, the possible functional consequences, and the potential effects on health.


Nature | 2018

Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes

Sebastian Mueller; Thomas Engleitner; Roman Maresch; Magdalena Zukowska; Sebastian Lange; Thorsten Kaltenbacher; Björn Konukiewitz; Rupert Öllinger; Maximilian Zwiebel; Alex Strong; Hsi-Yu Yen; Ruby Banerjee; Sandra Louzada; Beiyuan Fu; Barbara Seidler; Juliana Götzfried; Kathleen Schuck; Zonera Hassan; Andreas Arbeiter; Nina Schönhuber; Sabine Klein; Christian Veltkamp; Mathias Friedrich; Lena Rad; Maxim Barenboim; Christoph Ziegenhain; Julia Hess; Oliver M. Dovey; Stefan Eser; Swati Parekh

The poor correlation of mutational landscapes with phenotypes limits our understanding of the pathogenesis and metastasis of pancreatic ductal adenocarcinoma (PDAC). Here we show that oncogenic dosage-variation has a critical role in PDAC biology and phenotypic diversification. We find an increase in gene dosage of mutant KRAS in human PDAC precursors, which drives both early tumorigenesis and metastasis and thus rationalizes early PDAC dissemination. To overcome the limitations posed to gene dosage studies by the stromal richness of PDAC, we have developed large cell culture resources of metastatic mouse PDAC. Integration of cell culture genomes, transcriptomes and tumour phenotypes with functional studies and human data reveals additional widespread effects of oncogenic dosage variation on cell morphology and plasticity, histopathology and clinical outcome, with the highest KrasMUT levels underlying aggressive undifferentiated phenotypes. We also identify alternative oncogenic gains (Myc, Yap1 or Nfkb2), which collaborate with heterozygous KrasMUT in driving tumorigenesis, but have lower metastatic potential. Mechanistically, different oncogenic gains and dosages evolve along distinct evolutionary routes, licensed by defined allelic states and/or combinations of hallmark tumour suppressor alterations (Cdkn2a, Trp53, Tgfβ-pathway). Thus, evolutionary constraints and contingencies direct oncogenic dosage gain and variation along defined routes to drive the early progression of PDAC and shape its downstream biology. Our study uncovers universal principles of Ras-driven oncogenesis that have potential relevance beyond pancreatic cancer.


International Journal of Gynecological Cancer | 2011

Pegylated Liposomal Doxorubicin and Gemcitabine in a Fixed Dose Rate Infusion for the Treatment of Patients With Poor Prognosis of Recurrent Ovarian Cancer: A Phase Ib Study

Guillermo Crespo; Marta I. Sierra; Raquel Losa; Jose Pablo Berros; Noemi Villanueva; Joaquin Fra; Paula J. Fonseca; M. Luque; Yolanda Fernández; Pilar Blay; Miguel F. Sanmamed; C. Muriel; Emilio Esteban; A. J. Lacave

Introduction Pegylated liposomal doxorubicin (PLD) is currently the reference treatment for platinum-resistant ovarian cancer. The combination of PLD and gemcitabine and the administration of gemcitabine at a fixed dose rate infusion (FDRI) seem to have additive activity in this disease setting. We have launched a phase Ib study with the combination of FDRI gemcitabine followed by PLD in recurrent ovarian cancer with a platinum-free interval of less than 1 year, with parallel pharmacokinetic and pharmacogenetic studies. Methods The starting dose of gemcitabine was 1500 mg/m2, 10 mg/m2 per minute, every 2 weeks (±250 mg gemcitabine titration depending on toxicity), followed by PLD 35 mg/m2 every 4 weeks. Gemcitabine pharmacokinetics and equilibrative nucleoside transporter 1, deoxycytidine kinase, and ribonucleotide reductase M1 gene expression levels were studied. Results Thirty-five patients were treated at 3 different dose levels (DL). Dose level 1 was not tolerated. Nonfrail patients continued to be treated at DL-1 (G 1250 mg/m2 on day 1 and PLD 35 mg/m2 on days 1 and 15). Of 10 evaluable nonfrail patients, 4 displayed dose-limiting toxicity. Frail patients were treated at DL-2 (G 1250 mg/m2 on day 1 and PLD 35 mg/m2 on days 1 and 15). Of the 12 evaluable frail patients, 3 developed dose-limiting toxicity. Neutropenia, palmar-plantar erythrodysesthesia and stomatitis were the most common toxicities. The response rate was 42.8% (95% confidence interval [CI], 34.5%-51.1%), with 17.1% (6/35) complete responses and 25.7% (9/35) partial responses. The median progression-free survival was 7.7 months (95% CI, 2.2-13.1). The median overall survival was 13.9 months (95% CI, 9.4-18.4). The administration of PLD after gemcitabine did not influence gemcitabine pharmacokinetics or its metabolites. The addition of PLD to gemcitabine caused a larger and longer induction of the ribonucleotide reductase M1 gene. Patients with higher baseline levels of deoxycytidine kinase had longer progression-free survival. Conclusion The recommended dose for a phase II study of patients with recurrent ovarian cancer having poor prognosis is PLD, 35 mg/m2 on day 1, and gemcitabine, 1000 mg/m2 on days 1 and 15 delivered at an FDRI of 10 mg/m2 per minute in 28-day cycles.


Nanotoxicology | 2017

DNA methylation changes in human lung epithelia cells exposed to multi-walled carbon nanotubes

Marta I. Sierra; Laura Rubio; Gustavo F. Bayón; Isabel Cobo; Pablo Menendez; Paula Morales; Cristina Mangas; Rocío G. Urdinguio; Virginia Lopez; Adolfo Valdés; Gerard Vales; Ricard Marcos; Ramón Torrecillas; Agustín F. Fernández; Mario F. Fraga

Abstract Humans are increasingly exposed to nanoparticles and, although many of their physiological effects have been described, the molecular mechanisms underlying them are still largely unknown. The present study aimed to determine the possible role of certain epigenetic mechanisms in the cellular response of human lung epithelial cells that are triggered by long-term exposure to titanium dioxide nanoparticles (TiO2NPs) and multi-walled carbon nanotubes (MWCNTs). The results showed that exposure to TiO2NPs had only minor effects on genome-wide DNA methylation. However, we identified 755 CpG sites showing consistent DNA hypomethylation in cells exposed to MWCNTs. These sites were mainly located at low density CpG regions and enhancers, and very frequently on the X chromosome. Our results thus suggest that long-term MWCNT exposure may have important effects on the epigenome.


Oncotarget | 2018

Epigenetic dysregulation of TET2 in human glioblastoma

María G. García; Antonella Carella; Rocío G. Urdinguio; Gustavo F. Bayón; Virginia Lopez; Juan Ramon Tejedor; Marta I. Sierra; Estela García-Toraño; Pablo Santamarina; Raúl F Pérez; Cristina Mangas; Aurora Astudillo; M. Daniela Corte-Torres; Inés Sáenz-de-Santa-María; María-Dolores Chiara; Agustín F. Fernández; Mario F. Fraga

Ten-eleven translocation (TET) enzymes are frequently deregulated in cancer, but the underlying molecular mechanisms are still poorly understood. Here we report that TET2 shows frequent epigenetic alterations in human glioblastoma including DNA hypermethylation and hypo-hydroxymethylation, as well as loss of histone acetylation. Ectopic overexpression of TET2 regulated neural differentiation in glioblastoma cell lines and impaired tumor growth. Our results suggest that epigenetic dysregulation of TET2 plays a role in human glioblastoma.


The Journal of Clinical Endocrinology and Metabolism | 2018

SDHC Promoter Methylation, a Novel Pathogenic Mechanism in Parasympathetic Paragangliomas

Cristóbal Bernardo-Castiñeira; Nuria Valdés; Marta I. Sierra; Inés Sáenz-de-Santa-María; Gustavo F. Bayón; Raúl F Pérez; Agustín F. Fernández; Mario F. Fraga; Aurora Astudillo; Rafael Menéndez; Belén Fernández; Maribel del Olmo; Carlos Suarez; María-Dolores Chiara

Context Germline mutations in the succinate dehydrogenase A, B, C, and D genes (collectively, SDHx) predispose to the development of paragangliomas (PGLs) arising at the parasympathetic or sympathetic neuroendocrine systems. SDHx mutations cause absence of tumoral immunostaining for SDHB. However, negative SDHB immunostaining has also been found in a subset of PGLs that lack SDHx mutations. Settings Here, we report the comprehensive molecular characterization of one such a tumor of parasympathetic origin compared with healthy paraganglia and other PGLs with or without SDHx mutations. Results Integration of multiplatform data revealed somatic SDHC methylation and loss of the 1q23.3 region containing the SDHC gene. This correlated with decreased SDHC messenger RNA (mRNA) and protein levels. Furthermore, another genetic event found affected the VHL gene, which showed a decreased DNA copy number, associated with low VHL mRNA levels, and an absence of VHL protein detected by immunohistochemistry. In addition, the tumor displayed a pseudohypoxic phenotype consisting in overexpression of the hypoxia-inducible factor (HIF)-1α and miR-210, as well as downregulation of the iron-sulfur cluster assembly enzyme (ISCU) involved in SDHB maturation. This profile resembles that of SDHx- or VHL-mutated PGLs but not of PGLs with decreased VHL copy number, pointing to SDHC rather than VHL as the pathogenic driver. Conclusions Collectively, these findings demonstrate the potential importance of both the SDHC epigenomic event and the activation of the HIF-1α/miR-210/ISCU axis in the pathogenesis of SDHx wild-type/SDHB-negative PGLs. To our knowledge, this is the first case of a sporadic parasympathetic PGL that carries silencing of SDHC, fulfilling the two-hit Knudsons model for tumorigenesis.


Epigenetics | 2018

DNA methylation reprogramming of human cancer cells by expression of a plant 5-methylcytosine DNA glycosylase

Teresa Morales-Ruiz; María Victoria García-Ortiz; Iván Devesa-Guerra; Laura Raya-Ruiz; Juan Ramon Tejedor; Gustavo F. Bayón; Marta I. Sierra; Mario F. Fraga; Rafael R. Ariza; Teresa Roldán-Arjona

ABSTRACT Patterns of DNA methylation, an important epigenetic modification involved in gene silencing and development, are disrupted in cancer cells. Understanding the functional significance of aberrant methylation in tumors remains challenging, due in part to the lack of suitable tools to actively modify methylation patterns. DNA demethylation caused by mammalian DNA methyltransferase inhibitors is transient and replication-dependent, whereas that induced by TET enzymes involves oxidized 5mC derivatives that perform poorly understood regulatory functions. Unlike animals, plants possess enzymes that directly excise unoxidized 5mC from DNA, allowing restoration of unmethylated C through base excision repair. Here, we show that expression of Arabidopsis 5mC DNA glycosylase DEMETER (DME) in colon cancer cells demethylates and reactivates hypermethylated silenced loci. Interestingly, DME expression causes genome-wide changes that include both DNA methylation losses and gains, and partially restores the methylation pattern observed in normal tissue. Furthermore, such methylome reprogramming is accompanied by altered cell cycle responses and increased sensibility to anti-tumor drugs, decreased ability to form colonospheres, and tumor growth impairment in vivo. Our study shows that it is possible to reprogram a human cancer DNA methylome by expression of a plant DNA demethylase.

Collaboration


Dive into the Marta I. Sierra'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