Network


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

Hotspot


Dive into the research topics where Sílvia Xargay-Torrent is active.

Publication


Featured researches published by Sílvia Xargay-Torrent.


Blood | 2011

The Hsp90 inhibitor IPI-504 overcomes bortezomib resistance in mantle cell lymphoma in vitro and in vivo by down-regulation of the prosurvival ER chaperone BiP/Grp78

Gaël Roué; Patricia Pérez-Galán; Ana Mozos; Mónica López-Guerra; Sílvia Xargay-Torrent; Laia Rosich; Ifigènia Saborit-Villarroya; Emmanuel Normant; Elias Campo; Dolors Colomer

Despite the promising introduction of the proteasome inhibitor bortezomib in the treatment of mantle cell lymphoma (MCL), not all patients respond, and resistance often appears after initial treatment. By analyzing a set of 18 MCL samples, including cell lines with constitutive or induced resistance to bortezomib, we found a high correlation between loss of sensitivity to the proteasome inhibitor and up-regulation of the prosurvival chaperone BiP/Grp78. BiP/Grp78 stabilization was ensured at a posttranscriptional level by an increase in the chaperoning activity of heat shock protein of 90 kDa (Hsp90). In bortezomib-resistant cells, both BiP/Grp78 knockdown and cell pretreatment with the Hsp90 inhibitor of the ansamycin class, IPI-504, led to synergistic induction of apoptotic cell death when combined with bortezomib. Cell exposure to the IPI-504-bortezomib combination provoked the dissociation of Hsp90/BiP complexes, leading to BiP/Grp78 depletion, inhibition of unfolded protein response, and promotion of NOXA-mediated mitochondrial depolarization. The IPI-504-bortezomib combination also prevented BiP/Grp78 accumulation, thereby promoting apoptosis and inhibiting the growth of bortezomib-resistant tumors in a mouse model of MCL xenotransplantation. These results suggest that targeting unfolded protein response activation by the inhibition of Hsp90 may be an attractive model for the design of a new bortezomib-based combination therapy for MCL.


Clinical Cancer Research | 2011

Vorinostat-Induced Apoptosis in Mantle Cell Lymphoma Is Mediated by Acetylation of Proapoptotic BH3-Only Gene Promoters

Sílvia Xargay-Torrent; Mónica López-Guerra; Ifigènia Saborit-Villarroya; Laia Rosich; Elias Campo; Gaël Roué; Dolors Colomer

Purpose: Mantle cell lymphoma (MCL) is an aggressive B-cell neoplasm with generally poor prognosis, for which current therapies have shown limited efficacy. Vorinostat is a histone deacetylase inhibitor (HDACi) that has been approved for the treatment of cutaneous T-cell lymphoma. Our purpose was to describe the molecular mechanism whereby vorinostat induces apoptosis in MCL with particular emphasis on the role of proapoptotic BH3-only proteins. Experimental Design: The sensitivity to vorinostat was analyzed in eight MCL cell lines and primary cells from 10 MCL patients. Determination of vorinostat mechanism of action was done by flow cytometry, immunoblotting, HDAC activity assay kit, quantitative reverse transcription PCR, chromatin immunoprecipitation, and siRNA-mediated transfection. Results: Vorinostat inhibited total histone deacetylase activity leading to selective toxicity toward tumor cells. Vorinostat-mediated cell death implied the activation of mitochondrial apoptosis, as attested by BAX and BAK conformational changes, mitochondrial depolarization, reactive oxygen species generation, and subsequent caspase-dependent cell death. This phenomenon was linked to H4 hyperacetylation on promoter regions and consequent transcriptional activation of the proapoptotic BH3-only genes BIM, BMF, and NOXA. Selective knockdown of the three corresponding proteins rescued cells from vorinostat-induced apoptosis. Moreover, vorinostat enhanced the activity of the BH3-mimetic ABT-263 in MCL cells, leading to synergistic apoptosis induction. Conclusion: These results indicated that transcriptional upregulation of BH3-only proteins plays an important role in the antitumoral activity of vorinostat in MCL, and that HDACi alone or in combination with BH3-mimetizing agents may represent a promising therapeutic approach for MCL patients. Clin Cancer Res; 17(12); 3956–68. ©2011 AACR.


Clinical Cancer Research | 2012

Counteracting Autophagy Overcomes Resistance to Everolimus in Mantle Cell Lymphoma

Laia Rosich; Sílvia Xargay-Torrent; Mónica López-Guerra; Elias Campo; Dolors Colomer; Gaël Roué

Purpose: Mantle cell lymphoma (MCL) is an aggressive B-lymphoid neoplasm with poor response to conventional chemotherapy and short survival. The phosphatidylinositol 3-kinase/Akt/mTOR survival pathway is constitutively activated in MCL cells, thereby making the mTOR inhibition an attractive therapeutic strategy. The first clinical studies of everolimus (RAD001), an mTOR inhibitor, in relapsed MCL patients have reported a significant response. Our aim was to analyze the mechanism related to everolimus resistance/sensitivity in MCL cells. Experimental Design: Sensitivity to everolimus was analyzed in MCL cell lines and primary MCL cells. Everolimus mechanism of action was determined by flow cytometry and Western blot. Particularly, autophagy was studied by LC3BI/II expression, autophagolysosomes detection by flow cytometry and fluorescence microscopy, and siRNA-mediated gene silencing. Results: Everolimus exerted antitumoral effect on MCL cells while sparing normal cells. In MCL cell lines, this phenomenon was associated to G1 cell-cycle arrest, dephosphorylation of the mTOR downstream targets, 4E-BP1 and S6RP, and rephosphorylation of Akt. A synergistic cytotoxic effect was observed between everolimus and an Akt inhibitor, which overcame the compensatory reactivation within the mTOR signaling pathway. Interestingly, MCL cells with low response to this combination showed high levels of autophagy. Accordingly, selective triple knockdown of the autophagy genes ATG7, ATG5 and ATG3, and pretreatment with the autophagy inhibitor hydroxychloroquine, efficiently overcame the resistance to Akt/mTOR inhibitors, leading to the activation of the mitochondrial apoptotic pathway. Conclusions: These results suggest that autophagy induction protects MCL cells from Akt/mTOR targeting and counteracting autophagy may represent an attractive strategy for sensitizing MCL cells to everolimus-based therapy. Clin Cancer Res; 18(19); 5278–89. ©2012 AACR.


Leukemia | 2015

The γ-secretase inhibitor PF-03084014 combined with fludarabine antagonizes migration, invasion and angiogenesis in NOTCH1-mutated CLL cells

Mónica López-Guerra; Sílvia Xargay-Torrent; Laia Rosich; Arnau Montraveta; J Roldán; A Matas-Céspedes; Neus Villamor; M. Aymerich; Carlos López-Otín; Patricia Pérez-Galán; Gaël Roué; Elias Campo; Dolors Colomer

Targeting Notch signaling has emerged as a promising therapeutic strategy for chronic lymphocytic leukemia (CLL), especially for the poor prognostic subgroup of NOTCH1-mutated patients. Here, we report that the γ-secretase inhibitor PF-03084014 inhibits the constitutive Notch activation and induces selective apoptosis in CLL cells carrying NOTCH1 mutations. Combination of PF-03084014 with fludarabine has a synergistic antileukemic effect in primary NOTCH1-mutated CLL cells, even in the presence of the protective stroma. At transcriptional level, PF-03084014 plus fludarabine treatment induces the upregulation of the proapoptotic gene HRK and the downmodulation of MMP9, IL32 and RAC2 genes that are related to invasion and chemotaxis. PF-03084014 also overcomes fludarabine-mediated activation of nuclear factor-κB signaling. Moreover, this combination impairs angiogenesis and CXCL12-induced responses in NOTCH1-mutated CLL cells, in particular those related to tumoral migration and invasion. Importantly, all these collaborative effects are specific for NOTCH1 mutation and do not occur in unmutated cases. In conclusion, we provide evidence that Notch is a therapeutic target in CLL cases with NOTCH1-activating mutations, supporting the use of Notch pathway inhibitors in combination with chemotherapy as a promising approach for the treatment of these high-risk CLL patients.


Haematologica | 2013

The phosphatidylinositol-3-kinase inhibitor NVP-BKM120 overcomes resistance signals derived from microenvironment by regulating the Akt/FoxO3a/Bim axis in chronic lymphocytic leukemia cells

Laia Rosich; Ifigènia Saborit-Villarroya; Mónica López-Guerra; Sílvia Xargay-Torrent; Arnau Montraveta; Marta Aymerich; Neus Villamor; Elias Campo; Patricia Pérez-Galán; Gaël Roué; Dolors Colomer

Phosphatidylinositol-3-kinase pathway is constitutively activated in chronic lymphocytic leukemia mainly due to microenvironment signals, including stromal cell interaction and CXCR4 and B-cell receptor activation. Because of the importance of phosphatidylinositol-3-kinase signaling in chronic lymphocytic leukemia, we investigated the activity of the NVP-BKM120, an orally available pan class I phosphatidylinositol-3-kinase inhibitor. Sensitivity to NVP-BKM120 was analyzed in chronic lymphocytic leukemia primary samples in the context of B-cell receptor and microenvironment stimulation. NVP-BKM120 promoted mitochondrial apoptosis in most primary cells independently of common prognostic markers. NVP-BKM120 activity induced the blockage of phosphatidylinositol-3-kinase signaling, decreased Akt and FoxO3a phosphorylation leading to concomitant Mcl-1 downregulation and Bim induction. Accordingly, selective knockdown of BIM rescued cells from NVP-BKM120-induced apoptosis, while the kinase inhibitor synergistically enhanced the apoptosis induced by the BH3-mimetic ABT-263. We also found NVP-BKM120 to inhibit B-cell receptor- and stroma-dependent Akt pathway activation, thus sensitizing chronic lymphocytic leukemia cells to bendamustine and fludarabine. Furthermore, NVP-BKM120 down-regulated secretion of chemokines after B-cell receptor stimulation and inhibited cell chemotaxis and actin polymerization upon CXCR4 triggering by CXCL12. Our findings establish that NVP-BKM120 effectively inhibits the phosphatidylinositol-3-kinase signaling pathway and disturbs the protective effect of the tumor microenvironment with the subsequent apoptosis induction through the Akt/FoxO3a/Bim axis. We provide here a strong rationale for undertaking clinical trials of NVP-BKM120 in chronic lymphocytic leukemia patients alone or in combination therapies.


PLOS ONE | 2011

Epigenetic Activation of SOX11 in Lymphoid Neoplasms by Histone Modifications

Maria Carmela Vegliante; Cristina Royo; Jara Palomero; Itziar Salaverria; Balázs Bálint; Idoia Martin-Guerrero; Xabier Agirre; Amaia Lujambio; Julia Richter; Sílvia Xargay-Torrent; Sílvia Beà; Luis Hernández; Anna Enjuanes; María José Calasanz; Andreas Rosenwald; German Ott; Jose Roman-Gomez; Felipe Prosper; Manel Esteller; Pedro Jares; Reiner Siebert; Elias Campo; José I. Martín-Subero; Virginia Amador

Recent studies have shown aberrant expression of SOX11 in various types of aggressive B-cell neoplasms. To elucidate the molecular mechanisms leading to such deregulation, we performed a comprehensive SOX11 gene expression and epigenetic study in stem cells, normal hematopoietic cells and different lymphoid neoplasms. We observed that SOX11 expression is associated with unmethylated DNA and presence of activating histone marks (H3K9/14Ac and H3K4me3) in embryonic stem cells and some aggressive B-cell neoplasms. In contrast, adult stem cells, normal hematopoietic cells and other lymphoid neoplasms do not express SOX11. Such repression was associated with silencing histone marks H3K9me2 and H3K27me3. The SOX11 promoter of non-malignant cells was consistently unmethylated whereas lymphoid neoplasms with silenced SOX11 tended to acquire DNA hypermethylation. SOX11 silencing in cell lines was reversed by the histone deacetylase inhibitor SAHA but not by the DNA methyltransferase inhibitor AZA. These data indicate that, although DNA hypermethylation of SOX11 is frequent in lymphoid neoplasms, it seems to be functionally inert, as SOX11 is already silenced in the hematopoietic system. In contrast, the pathogenic role of SOX11 is associated with its de novo expression in some aggressive lymphoid malignancies, which is mediated by a shift from inactivating to activating histone modifications.


Clinical Cancer Research | 2013

Sorafenib inhibits cell migration and stroma-mediated bortezomib resistance by interfering B-cell receptor signaling and protein translation in mantle cell lymphoma

Sílvia Xargay-Torrent; Mónica López-Guerra; Arnau Montraveta; Ifigènia Saborit-Villarroya; Laia Rosich; Alba Navarro; Patricia Pérez-Galán; Gaël Roué; Elias Campo; Dolors Colomer

Purpose: We evaluated the antitumoral properties of the multikinase inhibitor sorafenib in mantle cell lymphoma (MCL), an aggressive B lymphoma for which current therapies have shown limited efficacy. Experimental Design: Sensitivity to sorafenib was analyzed in MCL cell lines and primary samples in the context of BCR and microenvironment simulation. Sorafenib signaling was characterized by quantitative PCR, Western blotting, immunofluorescence, and protein immunoprecipitation. Migration analysis included flow cytometric counting, actin polymerization assays, and siRNA-mediated knockdown of focal adhesion kinase (FAK). In vivo antitumor effect of sorafenib and bortezomib was analyzed in an MCL xenograft mouse model. Results: Sorafenib rapidly dephosphorylates the BCR-associated kinases, Syk and Lyn, as well as FAK, an Src target involved in focal adhesion. In this line, sorafenib displays strong synergy with the Syk inhibitor, R406. Sorafenib also blocks Mcl-1 and cyclin D1 translation, which promotes an imbalance between pro- and antiapoptotic proteins and facilitates Bax release from cyclin D1, leading to the induction of mitochondrial apoptosis and caspase-dependent and -independent mechanisms. Moreover, sorafenib inhibits MCL cell migration and CXCL12-induced actin polymerization. FAK knockdown partially prevents this inhibitory effect, indicating that FAK is a relevant target of sorafenib. Furthermore, sorafenib enhances the antitumoral activity of bortezomib in an MCL xenograft mouse model as well as overcomes stroma-mediated bortezomib resistance in MCL cells. Conclusion: We show for the first time that sorafenib interferes with BCR signaling, protein translation and modulates the microenvironment prosurvival signals in MCL, suggesting that sorafenib, alone or in combination with bortezomib, may represent a promising approach to treat patients with MCL. Clin Cancer Res; 19(3); 586–97. ©2012 AACR.


Leukemia | 2012

Sorafenib targets BCR kinases and blocks migratory and microenvironmental survival signals in CLL cells.

Mónica López-Guerra; Sílvia Xargay-Torrent; Patricia Pérez-Galán; Ifigènia Saborit-Villarroya; Laia Rosich; Neus Villamor; M. Aymerich; Gaël Roué; Elias Campo; Emilio Montserrat; Dolors Colomer

Sorafenib targets BCR kinases and blocks migratory and microenvironmental survival signals in CLL cells


Oncotarget | 2016

CD69 expression potentially predicts response to bendamustine and its modulation by ibrutinib or idelalisib enhances cytotoxic effect in chronic lymphocytic leukemia

Arnau Montraveta; Eriong Lee-Vergés; Jocabed Roldán; Laura Jiménez; Sandra Cabezas; Guillem Clot; Magda Pinyol; Sílvia Xargay-Torrent; Laia Rosich; Cristina Arimany-Nardi; Marta Aymerich; Neus Villamor; Armando López-Guillermo; Patricia Pérez-Galán; Gaël Roué; Marçal Pastor-Anglada; Elias Campo; Mónica López-Guerra; Dolors Colomer

Clinical responses to bendamustine in chronic lymphocytic leukemia (CLL) are highly heterogeneous and no specific markers to predict sensitivity to this drug have been reported. In order to identify biomarkers of response, we analyzed the in vitro activity of bendamustine and the gene expression profile in primary CLL cells. We observed that mRNA expression of CD69 (CD69) and ITGAM (CD11b) constitute the most powerful predictor of response to bendamustine. When we interrogated the predictive value of the corresponding cell surface proteins, the expression of the activation marker CD69 was the most reliable predictor of sensitivity to bendamustine. Importantly, a multivariate analysis revealed that the predictive value of CD69 expression was independent from other clinico-biological CLL features. We also showed that when CLL cells were co-cultured with distinct subtypes of stromal cells, an upregulation of CD69 was accompanied by a reduced sensitivity to bendamustine. In agreement with this, tumor cells derived from lymphoid tumor niches harbored higher CD69 expression and were less sensitive to bendamustine than their peripheral blood counterparts. Furthermore, pretreatment of CD69 high CLL cases with the B-cell receptor (BCR) pathway inhibitors ibrutinib and idelalisib decreased CD69 levels and enhanced bendamustine cytotoxic effect. Collectively, our findings indicate that CD69 could be a predictor of bendamustine response in CLL patients and the combination of clinically-tested BCR signaling inhibitors with bendamustine may represent a promising strategy for bendamustine low responsive CLL cases.


PLOS ONE | 2018

Circulating sex hormone binding globulin: An integrating biomarker for an adverse cardio-metabolic profile in obese pregnant women

Sílvia Xargay-Torrent; Gemma Carreras-Badosa; Sara Borrat-Padrosa; Anna Prats-Puig; Pilar Soriano; Elena Álvarez-Castaño; Mª Jose Ferri; Francis de Zegher; Lourdes Ibáñez; Abel López-Bermejo; Judit Bassols

Sex hormone-binding globulin (SHBG) negatively associates with pre-gestational body mass index (BMI) and gestational weight gain. The link with other cardio-metabolic risk factors in pregnant women is poorly understood. Our aim was to study the association of SHBG levels with common cardio-metabolic risk parameters in pregnant woman. Serum SHBG was quantified in 291 Caucasian pregnant women (142 with normal weight, 42 with pregestational obesity, 50 with gestational obesity and 57 with pregestational plus gestational obesity) with uncomplicated pregnancies and parturition. Cardio-metabolic [C-reactive protein (CRP), blood pressure (BP), glycosylated hemoglobin (HbAc1), glucose, C-peptide, insulin, triglycerides and high molecular weight (HMW) adiponectin], and endocrine [testosterone and estradiol] parameters were also assessed. SHBG was negatively correlated with BMI, but also with CRP, BP, HbAc1, pre and post-load glucose, C-peptide, HOMA-IR, triglycerides; and positively with HMW adiponectin (all p<0.01 to p<0.0001). These associations were more robust in women with pregestational plus gestational obesity, who had lower SHBG, in comparison to normal-weight women (p<0.0001). In multivariate analyses in women with pregestational plus gestational obesity SHBG showed independent associations with CRP (β = −0.352, p = 0.03, R2 = 8.0%), DBP (β = −0.353, p = 0.03, R2 = 7.0%) and SBP (β = −0.333, p = 0.04, R2 = 6.0%) independently of BMI and metabolic and endocrine parameters. SHBG is decreased in pregnant women with pregestational plus gestational obesity in association with common cardio-metabolic parameters. SHBG could represent an integrating biomarker for an adverse cardio-metabolic profile in pregnant women with pregestational plus gestational obesity.

Collaboration


Dive into the Sílvia Xargay-Torrent's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elias Campo

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Laia Rosich

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Gaël Roué

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge