Network


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

Hotspot


Dive into the research topics where Pilar Santisteban is active.

Publication


Featured researches published by Pilar Santisteban.


JAMA | 2013

Association Between BRAF V600E Mutation and Mortality in Patients With Papillary Thyroid Cancer

Mingzhao Xing; Ali S. Alzahrani; Kathryn A. Carson; David Viola; Rossella Elisei; Bela Bendlova; Linwah Yip; Caterina Mian; Federica Vianello; R. Michael Tuttle; Eyal Robenshtok; James A. Fagin; Efisio Puxeddu; Laura Fugazzola; Agnieszka Czarniecka; Barbara Jarzab; Christine J. O’Neill; Mark S. Sywak; Alfred King-Yin Lam; Garcilaso Riesco-Eizaguirre; Pilar Santisteban; Hirotaka Nakayama; Ralph P. Tufano; Sara I. Pai; Martha A. Zeiger; William H. Westra; Douglas P. Clark; Roderick J. Clifton-Bligh; David Sidransky; Paul W. Ladenson

IMPORTANCE BRAF V600E is a prominent oncogene in papillary thyroid cancer (PTC), but its role in PTC-related patient mortality has not been established. OBJECTIVE To investigate the relationship between BRAF V600E mutation and PTC-related mortality. DESIGN, SETTING, AND PARTICIPANTS Retrospective study of 1849 patients (1411 women and 438 men) with a median age of 46 years (interquartile range, 34-58 years) and an overall median follow-up time of 33 months (interquartile range, 13-67 months) after initial treatment at 13 centers in 7 countries between 1978 and 2011. MAIN OUTCOMES AND MEASURES Patient deaths specifically caused by PTC. RESULTS Overall, mortality was 5.3% (45/845; 95% CI, 3.9%-7.1%) vs 1.1% (11/1004; 95% CI, 0.5%-2.0%) (P < .001) in BRAF V600E-positive vs mutation-negative patients. Deaths per 1000 person-years in the analysis of all PTC were 12.87 (95% CI, 9.61-17.24) vs 2.52 (95% CI, 1.40-4.55) in BRAF V600E-positive vs mutation-negative patients; the hazard ratio (HR) was 2.66 (95% CI, 1.30-5.43) after adjustment for age at diagnosis, sex, and medical center. Deaths per 1000 person-years in the analysis of the conventional variant of PTC were 11.80 (95% CI, 8.39-16.60) vs 2.25 (95% CI, 1.01-5.00) in BRAF V600E-positive vs mutation-negative patients; the adjusted HR was 3.53 (95% CI, 1.25-9.98). When lymph node metastasis, extrathyroidal invasion, and distant metastasis were also included in the model, the association of BRAF V600E with mortality for all PTC was no longer significant (HR, 1.21; 95% CI, 0.53-2.76). A higher BRAF V600E-associated patient mortality was also observed in several clinicopathological subcategories, but statistical significance was lost with adjustment for patient age, sex, and medical center. For example, in patients with lymph node metastasis, the deaths per 1000 person-years were 26.26 (95% CI, 19.18-35.94) vs 5.93 (95% CI, 2.96-11.86) in BRAF V600E-positive vs mutation-negative patients (unadjusted HR, 4.43 [95% CI, 2.06-9.51]; adjusted HR, 1.46 [95% CI, 0.62-3.47]). In patients with distant tumor metastasis, deaths per 1000 person-years were 87.72 (95% CI, 62.68-122.77) vs 32.28 (95% CI, 16.14-64.55) in BRAF V600E-positive vs mutation-negative patients (unadjusted HR, 2.63 [95% CI, 1.21-5.72]; adjusted HR, 0.84 [95% CI, 0.27-2.62]). CONCLUSIONS AND RELEVANCE In this retrospective multicenter study, the presence of the BRAF V600E mutation was significantly associated with increased cancer-related mortality among patients with PTC. Because overall mortality in PTC is low and the association was not independent of tumor features, how to use BRAF V600E to manage mortality risk in patients with PTC is unclear. These findings support further investigation of the prognostic and therapeutic implications of BRAF V600E status in PTC.


Journal of Clinical Oncology | 2015

Association Between BRAF V600E Mutation and Recurrence of Papillary Thyroid Cancer

Mingzhao Xing; Ali S. Alzahrani; Kathryn A. Carson; Young Kee Shong; Tae Yong Kim; David Viola; Rossella Elisei; Bela Bendlova; Linwah Yip; Caterina Mian; Federica Vianello; R. Michael Tuttle; Eyal Robenshtok; James A. Fagin; Efisio Puxeddu; Laura Fugazzola; Agnieszka Czarniecka; Barbara Jarzab; Christine J. O'Neill; Mark S. Sywak; Alfred King-Yin Lam; Garcilaso Riesco-Eizaguirre; Pilar Santisteban; Hirotaka Nakayama; Roderick J. Clifton-Bligh; Giovanni Tallini; Elizabeth H. Holt; Vlasta Sýkorová

PURPOSE To investigate the prognostic value of BRAF V600E mutation for the recurrence of papillary thyroid cancer (PTC). PATIENTS AND METHODS This was a retrospective multicenter study of the relationship between BRAF V600E mutation and recurrence of PTC in 2,099 patients (1,615 women and 484 men), with a median age of 45 years (interquartile range [IQR], 34 to 58 years) and a median follow-up time of 36 months (IQR, 14 to 75 months). RESULTS The overall BRAF V600E mutation prevalence was 48.5% (1,017 of 2,099). PTC recurrence occurred in 20.9% (213 of 1,017) of BRAF V600E mutation-positive and 11.6% (125 of 1,082) of BRAF V600E mutation-negative patients. Recurrence rates were 47.71 (95% CI, 41.72 to 54.57) versus 26.03 (95% CI, 21.85 to 31.02) per 1,000 person-years in BRAF mutation-positive versus -negative patients (P < .001), with a hazard ratio (HR) of 1.82 (95% CI, 1.46 to 2.28), which remained significant in a multivariable model adjusting for patient sex and age at diagnosis, medical center, and various conventional pathologic factors. Significant association between BRAF mutation and PTC recurrence was also found in patients with conventionally low-risk disease stage I or II and micro-PTC and within various subtypes of PTC. For example, in BRAF mutation-positive versus -negative follicular-variant PTC, recurrence occurred in 21.3% (19 of 89) and 7.0% (24 of 342) of patients, respectively, with recurrence rates of 53.84 (95% CI, 34.34 to 84.40) versus 19.47 (95% CI, 13.05 to 29.04) per 1,000 person-years (P < .001) and an HR of 3.20 (95% CI, 1.46 to 7.02) after adjustment for clinicopathologic factors. BRAF mutation was associated with poorer recurrence-free probability in Kaplan-Meier survival analyses in various clinicopathologic categories. CONCLUSION This large multicenter study demonstrates an independent prognostic value of BRAF V600E mutation for PTC recurrence in various clinicopathologic categories.


Cancer Research | 2009

The BRAFV600E Oncogene Induces Transforming Growth Factor β Secretion Leading to Sodium Iodide Symporter Repression and Increased Malignancy in Thyroid Cancer

Garcilaso Riesco-Eizaguirre; Irene Rodríguez; Antonio De la Vieja; Eugenia Costamagna; Nancy Carrasco; Manuel Nistal; Pilar Santisteban

The activating mutation BRAF(V600E) is a frequent genetic event in papillary thyroid carcinomas (PTC) that predicts a poor prognosis, leading to loss of sodium/iodide symporter (NIS) expression and subsequent radioiodide-refractory metastatic disease. The molecular basis of such an aggressive behavior induced by BRAF remains unclear. Here, we show a mechanism through which BRAF induces NIS repression and promotes epithelial to mesenchimal transition and invasion based on the operation of an autocrine transforming growth factor (TGF)beta loop. BRAF induces secretion of functional TGFbeta and blocking TGFbeta/Smad signaling at multiple levels rescues BRAF-induced NIS repression. Although this mechanism is MAP/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK independent, secreted TGFbeta cooperates with MEK-ERK signaling in BRAF-induced cell migration, Matrigel invasion, and EMT. Consistent with this process, TGFbeta and other key components of TGFbeta signaling, such as TbetaRII and pSmad2, are overexpressed in human PTC, suggesting a widespread activation of this pathway by locally released TGFbeta. Moreover, this high TGFbeta/Smad activity is associated with PTC invasion, nodal metastasis, and BRAF status. Interestingly, TGFbeta is overexpressed in the invasive front, whereas NIS is preferentially expressed in the central regions of the tumors, suggesting that this negative correlation between TGFbeta and NIS occurs locally inside the tumor. Our study describes a novel mechanism of NIS repression in thyroid cancer and provides evidence that TGFbeta may play a key role in promoting radioiodide resistance and tumor invasion during PTC progression.


PLOS Genetics | 2009

The Variant rs1867277 in FOXE1 Gene Confers Thyroid Cancer Susceptibility through the Recruitment of USF1/USF2 Transcription Factors

Iñigo Landa; Sergio Ruiz-Llorente; Cristina Montero-Conde; Lucía Inglada-Pérez; Francesca Schiavi; Susanna Leskelä; Guillermo Pita; Roger L. Milne; Javier Maravall; Ignacio Ramos; Víctor Andía; Paloma Rodríguez-Poyo; Antonino Jara-Albarrán; Amparo Meoro; Cristina Del Peso; Luis Arribas; Pedro Iglesias; Javier Caballero; Joaquín Serrano; Antonio Picó; Francisco Pomares; Gabriel Giménez; Pedro López-Mondéjar; Roberto Castello; Isabella Merante-Boschin; Maria Rosa Pelizzo; Didac Mauricio; Giuseppe Opocher; Cristina Rodríguez-Antona; Anna González-Neira

In order to identify genetic factors related to thyroid cancer susceptibility, we adopted a candidate gene approach. We studied tag- and putative functional SNPs in genes involved in thyroid cell differentiation and proliferation, and in genes found to be differentially expressed in thyroid carcinoma. A total of 768 SNPs in 97 genes were genotyped in a Spanish series of 615 cases and 525 controls, the former comprising the largest collection of patients with this pathology from a single population studied to date. SNPs in an LD block spanning the entire FOXE1 gene showed the strongest evidence of association with papillary thyroid carcinoma susceptibility. This association was validated in a second stage of the study that included an independent Italian series of 482 patients and 532 controls. The strongest association results were observed for rs1867277 (OR[per-allele] = 1.49; 95%CI = 1.30–1.70; P = 5.9×10−9). Functional assays of rs1867277 (NM_004473.3:c.−283G>A) within the FOXE1 5′ UTR suggested that this variant affects FOXE1 transcription. DNA-binding assays demonstrated that, exclusively, the sequence containing the A allele recruited the USF1/USF2 transcription factors, while both alleles formed a complex in which DREAM/CREB/αCREM participated. Transfection studies showed an allele-dependent transcriptional regulation of FOXE1. We propose a FOXE1 regulation model dependent on the rs1867277 genotype, indicating that this SNP is a causal variant in thyroid cancer susceptibility. Our results constitute the first functional explanation for an association identified by a GWAS and thereby elucidate a mechanism of thyroid cancer susceptibility. They also attest to the efficacy of candidate gene approaches in the GWAS era.


Oncogene | 2004

The small GTP-binding protein, Rhes, regulates signal transduction from G protein-coupled receptors

Pierfrancesco Vargiu; Ricardo de Abajo; Juan Antonio Garcia-Ranea; Alfonso Valencia; Pilar Santisteban; Piero Crespo; Juan Bernal

The Ras homolog enriched in striatum, Rhes, is the product of a thyroid hormone-regulated gene during brain development. Rhes and the dexamethasone-induced Dexras1 define a novel distinct subfamily of proteins within the Ras family, characterized by an extended variable domain in the carboxyl terminal region. We have carried this study because there is a complete lack of knowledge on Rhes signaling. We show that in PC12 cells, Rhes is targeted to the plasma membrane by farnesylation. We demonstrate that about 30% of the native Rhes protein is bound to GTP and this proportion is unaltered by typical Ras family nucleotide exchange factors. However, Rhes is not transforming in murine fibroblasts. We have also examined the role of Rhes in cell signaling. Rhes does not stimulate the ERK pathway. By contrast, it binds to and activates PI3K. On the other hand, we demonstrate that Rhes impairs the activation of the cAMP/PKA pathway by thyroid-stimulating hormone, and by an activated β2 adrenergic receptor by a mechanism that suggests uncoupling of the receptor to its cognate heterotrimeric complex. Overall, our results provide the initial insights into the role in signal transduction of this novel Ras family member.


Molecular and Cellular Endocrinology | 2003

TSH-activated signaling pathways in thyroid tumorigenesis

Marcos Rivas; Pilar Santisteban

Thyrotropin (TSH) is considered the main regulator of thyrocyte differentiation and proliferation. Thus, the characterization of the different signaling pathways triggered by TSH on these cells is of major interest in order to understand the mechanisms implicated in thyroid pathology. In this review we focus on the different signaling pathways involved in TSH-mediated proliferation and their role in thyroid transformation and tumorigenesis. TSH mitogenic activities are mediated largely by cAMP, which in turn may activate protein kinase (PKA)-dependent and independent processes. We analyze the effects of increased cAMP levels and PKA activity during cell cycle progression and the role of this signaling pathway in thyroid tumor initiation. Alternative pathways to PKA in the cAMP-mediated proliferation appear to involve the small GTPases Rap1 and Ras. We analyze the Ras effectors (PI3K, RalGDS and Raf) that are thought to mediate its oncogenic activity, as well as the ability of Ras to induce apoptosis in thyrocytes. Finally, we discuss the activation of the PLC/PKC cascade by TSH in thyroid cells and the role of this signaling pathway in the TSH-mediated proliferation and tumorigenesis.


Biochimie | 1999

Thyroid-specific gene expression in the multi-step process of thyroid carcinogenesis.

Purificación Ros; Dévora L. Rossi; Alvaro Acebrón; Pilar Santisteban

Thyroid-specific transcription factors TTF-1 and Pax-8 play a decisive role in the determination and maintenance of cellular phenotype activating thyroglobulin (Tg), thyroperoxidase (TPO), thyrotropin receptor (TSH-R) and the sodium/iodide symporter (NIS) gene transcription. In the present work, we have studied the expression of TTF-1 and Pax-8 and their target genes in samples derived from thyroid neoplasms of follicular origin, as well as in medullary carcinoma (MTC), obtained from surgery or from fine needle aspiration (FNA) biopsies. The results show that TTF-1 and Pax-8 are expressed in well differentiated adenomas and that their expression decreases in less differentiated papillary and follicular carcinomas and is lost in undifferentiated anaplastic carcinomas. Parallel levels of Tg, TPO and TSH-R expression were found in the same neoplasm samples. Interestingly TSH-R and TTF-1 gene expression was found in MTC samples. Furthermore, the expression of the thyroid-specific genes and their transcription factors is lost in thyroid cells derived from follicular, papillary and anaplastic human carcinomas. In these cells, Tg, TPO and TSH-R promoter activities were absent. Cotransfection with expression vectors for TTF-1 and Pax-8 resulted in the stimulation of transcription to a different extent for each promoter. These results may be clinically relevant for the evaluation and prognosis of thyroid cancer since the loss of specific markers correlates with the degree of tumor differentiation.


Journal of Neurochemistry | 2002

Ontogeny and Cellular Localization of the Pyruvate Recycling System in Rat Brain

Fátima Cruz; Steve R. Scott; Isabel Barroso; Pilar Santisteban; Sebastián Cerdán

Abstract: The ontogeny of the cerebral pyruvate recycling pathway and the cellular localization of associated enzymes, malic enzyme (ME) and phosphoenolpyruvate carboxykinase (PEPCK), have been investigated using a combination of 13C NMR spectroscopy, enzymatic analysis, and molecular biology approaches. Activity of the pathway, using [1,2‐13C2]acetate as a substrate, was detected by 13C NMR in brain extracts 3 weeks after birth, increasing progressively up to the third month of age. In whole‐brain homogenates, ME activity increased to adult levels with the same time course as the recycling pathway. PEPCK activity was low during the first 2 weeks of life and decreased further toward adulthood. ME and PEPCK activity were found in primary cultures of astrocytes and in synaptosomal fractions of adult brain. Primary cultures of cortical neurons showed PEPCK activity but no detectable ME activity. The cytosolic ME gene was expressed in primary cultures of neurons and in astrocytes as well as in the neonatal and adult brain. The PEPCK gene was expressed both in primary cultures of cortical neurons and in astrocytes, but the level of its expression in the neonatal and adult brain was undetectable.


Arquivos Brasileiros De Endocrinologia E Metabologia | 2007

TSH signalling and cancer

Custodia García-Jiménez; Pilar Santisteban

Thyroid cancers are the most frequent endocrine neoplasms and mutations in the thyrotropin receptor (TSHR) are unusually frequent. Here we present the state-of-the-art concerning the role of TSHR in thyroid cancer and discuss it in light of the cancer stem cell theory or the classical view. We briefly review the gene and protein structure updating the cancer related TSHR mutations database. Intriguingly, hyperfunctioning TSHR mutants characterise differentiated cancers in contrast to undifferentiated thyroid cancers which very often bear silenced TSHR. It remains unclear whether TSHR alterations in thyroid cancers play a role in the onset or they appear as a consequence of genetic instability during evolution, but the presence of functional TSHR is exploited in therapy. We outline the signalling network build up in the thyrocyte between TSHR/PKA and other proliferative pathways such as Wnt, PI3K and MAPK. This networks integrity surely plays a role in the onset/evolution of thyroid cancer and needs further research. Lastly, future investigation of epigenetic events occurring at the TSHR and other loci may give better clues for molecular based therapy of undifferentiated thyroid carcinomas. Targeted demethylating agents, histone deacetylase inhibitors combined with retinoids and specific RNAis may help treatment in the future.


Reviews in Endocrine & Metabolic Disorders | 2005

Thyroid development and effect on the nervous system

Pilar Santisteban; Juan Bernal

Supported by grants BFI2002-00489 (MCYT), BFU2004-03169 (MEC), GR/SAL/0382/2004 and GR/SAL/0773/2004 from Comunidad de Madrid and FIS, Instituto de Salud Carlos III, Red de Centros RCMN (C03/08), RCGC(C03/10), de gruposRGDM(G03/212) y PI041216 (Spain).

Collaboration


Dive into the Pilar Santisteban's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Evelyn F. Grollman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana Sastre-Perona

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Luis Lamas

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Antonio De la Vieja

Instituto de Salud Carlos III

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge