Network


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

Hotspot


Dive into the research topics where Sofia Torres is active.

Publication


Featured researches published by Sofia Torres.


Journal of Clinical Investigation | 2009

Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells

María Salazar; Arkaitz Carracedo; Íñigo J. Salanueva; Sonia Hernández-Tiedra; Mar Lorente; Ainara Egia; Patricia Vázquez; Cristina Blázquez; Sofia Torres; Stéphane Garcia; Jonathan Nowak; Gian Maria Fimia; Mauro Piacentini; Francesco Cecconi; Pier Paolo Pandolfi; Luis González-Feria; Juan L. Iovanna; Manuel Guzmán; Patricia Boya; Guillermo Velasco

Autophagy can promote cell survival or cell death, but the molecular basis underlying its dual role in cancer remains obscure. Here we demonstrate that delta(9)-tetrahydrocannabinol (THC), the main active component of marijuana, induces human glioma cell death through stimulation of autophagy. Our data indicate that THC induced ceramide accumulation and eukaryotic translation initiation factor 2alpha (eIF2alpha) phosphorylation and thereby activated an ER stress response that promoted autophagy via tribbles homolog 3-dependent (TRB3-dependent) inhibition of the Akt/mammalian target of rapamycin complex 1 (mTORC1) axis. We also showed that autophagy is upstream of apoptosis in cannabinoid-induced human and mouse cancer cell death and that activation of this pathway was necessary for the antitumor action of cannabinoids in vivo. These findings describe a mechanism by which THC can promote the autophagic death of human and mouse cancer cells and provide evidence that cannabinoid administration may be an effective therapeutic strategy for targeting human cancers.


Molecular Cancer Therapeutics | 2011

A combined preclinical therapy of cannabinoids and temozolomide against glioma.

Sofia Torres; Mar Lorente; Fátima Rodríguez-Fornés; Sonia Hernández-Tiedra; María Salazar; Elena García-Taboada; Juan Barcia; Manuel Guzmán; Guillermo Velasco

Glioblastoma multiforme (GBM) is highly resistant to current anticancer treatments, which makes it crucial to find new therapeutic strategies aimed at improving the poor prognosis of patients suffering from this disease. Δ9-Tetrahydrocannabinol (THC), the major active ingredient of marijuana, and other cannabinoid receptor agonists inhibit tumor growth in animal models of cancer, including glioma, an effect that relies, at least in part, on the stimulation of autophagy-mediated apoptosis in tumor cells. Here, we show that the combined administration of THC and temozolomide (TMZ; the benchmark agent for the management of GBM) exerts a strong antitumoral action in glioma xenografts, an effect that is also observed in tumors that are resistant to TMZ treatment. Combined administration of THC and TMZ enhanced autophagy, whereas pharmacologic or genetic inhibition of this process prevented TMZ + THC-induced cell death, supporting that activation of autophagy plays a crucial role on the mechanism of action of this drug combination. Administration of submaximal doses of THC and cannabidiol (CBD; another plant-derived cannabinoid that also induces glioma cell death through a mechanism of action different from that of THC) remarkably reduces the growth of glioma xenografts. Moreover, treatment with TMZ and submaximal doses of THC and CBD produced a strong antitumoral action in both TMZ-sensitive and TMZ-resistant tumors. Altogether, our findings support that the combined administration of TMZ and cannabinoids could be therapeutically exploited for the management of GBM. Mol Cancer Ther; 10(1); 90–103. ©2011 AACR.


Cell Death & Differentiation | 2011

Stimulation of the midkine/ALK axis renders glioma cells resistant to cannabinoid antitumoral action

Mar Lorente; Sofia Torres; María Salazar; Arkaitz Carracedo; Sonia Hernández-Tiedra; Fátima Rodríguez-Fornés; Elena García-Taboada; Bárbara Meléndez; Manuela Mollejo; Yolanda Campos-Martín; S A Lakatosh; Juan Barcia; Manuel Guzmán; Guillermo Velasco

Identifying the molecular mechanisms responsible for the resistance of gliomas to anticancer treatments is an issue of great therapeutic interest. Δ9-Tetrahydrocannabinol (THC), the major active ingredient of marijuana, and other cannabinoids inhibit tumor growth in animal models of cancer, including glioma, an effect that relies, at least in part, on the stimulation of autophagy-mediated apoptosis in tumor cells. Here, by analyzing the gene expression profile of a large series of human glioma cells with different sensitivity to cannabinoid action, we have identified a subset of genes specifically associated to THC resistance. One of these genes, namely that encoding the growth factor midkine (Mdk), is directly involved in the resistance of glioma cells to cannabinoid treatment. We also show that Mdk mediates its protective effect via the anaplastic lymphoma kinase (ALK) receptor and that Mdk signaling through ALK interferes with cannabinoid-induced autophagic cell death. Furthermore, in vivo Mdk silencing or ALK pharmacological inhibition sensitizes cannabinod-resistant tumors to THC antitumoral action. Altogether, our findings identify Mdk as a pivotal factor involved in the resistance of glioma cells to THC pro-autophagic and antitumoral action, and suggest that selective targeting of the Mdk/ALK axis could help to improve the efficacy of antitumoral therapies for gliomas.


Autophagy | 2009

TRB3 links ER stress to autophagy in cannabinoid anti-tumoral action

María Salazar; Arkaitz Carracedo; Íñigo J. Salanueva; Sonia Hernández-Tiedra; Ainara Egia; Mar Lorente; Patricia Vázquez; Sofia Torres; Juan L. Iovanna; Manuel Guzmán; Patricia Boya; Guillermo Velasco

Δ9-tetrahydrocannabinol (THC), the main active component of marijuana, is being investigated as a potential anti-tumoral agent. We find that THC stimulates an endoplasmic reticulum (ER) stress-related signaling pathway, which activates autophagy via inhibition of the Akt/mTORC1 axis. We also show that autophagy is upstream of apoptosis in cannabinoid-induced cancer cell death and that activation of this pathway is necessary for the anti-tumoral action of cannabinoids in vivo.


PLOS ONE | 2013

Local Delivery of Cannabinoid-Loaded Microparticles Inhibits Tumor Growth in a Murine Xenograft Model of Glioblastoma Multiforme

Dolores Hernán Pérez de la Ossa; Mar Lorente; Maria Esther Gil-Alegre; Sofia Torres; Elena García-Taboada; M.R. Aberturas; J. Molpeceres; Guillermo Velasco; Ana Isabel Torres-Suárez

Cannabinoids, the active components of marijuana and their derivatives, are currently investigated due to their potential therapeutic application for the management of many different diseases, including cancer. Specifically, Δ9-Tetrahydrocannabinol (THC) and Cannabidiol (CBD) – the two major ingredients of marijuana – have been shown to inhibit tumor growth in a number of animal models of cancer, including glioma. Although there are several pharmaceutical preparations that permit the oral administration of THC or its analogue nabilone or the oromucosal delivery of a THC- and CBD-enriched cannabis extract, the systemic administration of cannabinoids has several limitations in part derived from the high lipophilicity exhibited by these compounds. In this work we analyzed CBD- and THC-loaded poly-ε-caprolactone microparticles as an alternative delivery system for long-term cannabinoid administration in a murine xenograft model of glioma. In vitro characterization of THC- and CBD-loaded microparticles showed that this method of microencapsulation facilitates a sustained release of the two cannabinoids for several days. Local administration of THC-, CBD- or a mixture (1∶1 w:w) of THC- and CBD-loaded microparticles every 5 days to mice bearing glioma xenografts reduced tumour growth with the same efficacy than a daily local administration of the equivalent amount of those cannabinoids in solution. Moreover, treatment with cannabinoid-loaded microparticles enhanced apoptosis and decreased cell proliferation and angiogenesis in these tumours. Our findings support that THC- and CBD-loaded microparticles could be used as an alternative method of cannabinoid delivery in anticancer therapies.


Glia | 2009

Amphiregulin is a factor for resistance of glioma cells to cannabinoid-induced apoptosis.

Mar Lorente; Arkaitz Carracedo; Sofia Torres; Francesco Natali; Ainara Egia; Sonia Hernández-Tiedra; María Salazar; Cristina Blázquez; Manuel Guzmán; Guillermo Velasco

Gliomas, one of the most malignant forms of cancer, exhibit high resistance to conventional therapies. Identification of the molecular mechanisms responsible for this resistance is therefore of great interest to improve the efficacy of the treatments against these tumors. Δ9‐Tetrahydrocannabinol (THC), the major active ingredient of marijuana, and other cannabinoids inhibit tumor growth in animal models of cancer, including glioma, an effect that relies, at least in part, on the ability of these compounds to induce apoptosis of tumor cells. By analyzing the gene expression profile of two sub‐clones of C6 glioma cells with different sensitivity to cannabinoid‐induced apoptosis, we found a subset of genes with a marked differential expression in the two sub‐clones. Furthermore, we identified the epidermal growth factor receptor ligand amphiregulin as a candidate factor to mediate the resistance of glioma cells to cannabinoid treatment. Amphiregulin was highly overexpressed in the cannabinoid‐resistant cell line, both in culture and in tumor xenografts. Moreover, in vivo silencing of amphiregulin rendered the resistant tumors xenografts sensitive to cannabinoid antitumoral action. Amphiregulin expression was associated with increased extracellular signal‐regulated kinase (ERK) activation, which mediated the resistance to THC by blunting the expression of p8 and TRB3—two genes involved in cannabinoid‐induced apoptosis of glioma cells. Our findings therefore identify Amphirregulin as a factor for resistance of glioma cells to THC‐induced apoptosis and contribute to unraveling the molecular bases underlying the emerging notion that targeted inhibition of the EGFR pathway can improve the efficacy of antitumoral therapies.


Methods in Enzymology | 2011

Detecting autophagy in response to ER stress signals in cancer.

María Salazar; Sonia Hernández-Tiedra; Sofia Torres; Mar Lorente; Manuel Guzmán; Guillermo Velasco

Different physiological and pathological situations that produce alterations in the endoplasmic reticulum, lead to a condition known as ER stress. ER stress activates a complex intracellular signal transduction pathway, called unfolded protein response (UPR). UPR is tailored essentially to reestablish ER homeostasis. However, when persistent, ER stress can switch the cytoprotective functions of UPR into cell death promoting mechanisms. One of the cellular mechanisms that are regulated by ER stress is autophagy. Autophagy is a cellular process by which different cytoplasmic components including organelles are targeted for degradation to the autophagosomes. Interestingly, like ER stress, autophagy can be a protective or a cell death promoting mechanism. Recently, a variety of anticancer therapies (including those that stimulate ER stress) have been shown to activate autophagy in tumor cells, which has been proposed to either enhance cancer cell death or act as a mechanism of resistance to chemotherapy. In this chapter, we will describe some of the procedures that are currently used to analyze autophagy as well as some of the experimental approaches that can be undertaken to investigate the connection between ER stress and autophagy in cancer.


Archive | 2011

Copy Number Alterations in Glioma Cell Lines

Bárbara Meléndez; Ainoha García-Claver; Yolanda Ruano; Yolanda Campos-Martín; Ángel Rodríguez de Lope; Elisa Pérez-Magán; Pilar Mur; Sofia Torres; Mar Lorente; Guillermo Velasco; Manuela Mollejo

Established tumor-derived cell lines are widely and routinely used as in vitro cancer models for various kinds of biomedical research. The easy management of these cell cultures, in contrast to the inherent difficulty in establishing and mantaining primary tumoral cultures, has contributed to the wide use of these inmortalized cell lines in order to characterize the biological significance of specific genomic aberrations identified in primary tumors. Therefore, it has been assumed that the genomic and expression aberrations of long-term established cell lines resemble, and are representative, of the primary tumor from which the cell line was derived. Indeed, the cell line-based research has been performed, not only for the definition of the molecular biology of several cancer models, but also for the investigation of new targeted therapeutic agents in a prior step to clinical practice. The use of tumor-derived cell lines has been highly relevant for the testing and development of new therapeutical agents, with several cancer cell-line panels having been developed for drug sensitivity screening and new agents’ discovery (Sharma et al, 2010). Controversial concerning the ability of tumor-derived cell lines to accurately reflect the phenotype and genotype of the parental histology has been documented. A previous report of Greshock and coworkers using array-based Comparative Genomic Hybridization (aCGH) data of seven diagnosis-specific matched tumors and cell lines showed that, on average, cell lines preserve in vitro the genetic aberrations that are unique to the parent histology from which they were derived, while acquiring additional locus-specific alterations in long-term cultures (Greshock et al, 2007). In contrast, a study on breast cancer cell lines and primary tumors highlight that cell lines do not always represent the genotypes of parental tumor tissues (Tsuji et al, 2010). Furthermore, a parallel genomic and expression study on glioma cell lines and primary tumors states that in this specific cancer type, cell lines are poor representative of the primary tumors (Li et al, 2008). Given the importance of the use of cell lines as models for the study of the biology and development of tumors, and for the testing of the mode of action of new therapeutical agents, the knowledge of which genomic alterations are tumor-specific or which are necessary for the maintenance of the cell line in culture, becomes essential.


Biochemical Pharmacology | 2018

Targeting Glioma Initiating Cells With A Combined Therapy Of Cannabinoids And Temozolomide.

Israel López-Valero; Cristina Saiz-Ladera; Sofia Torres; Sonia Hernández-Tiedra; Elena García-Taboada; Fátima Rodríguez-Fornés; Marina Barba; David Dávila; Nélida Salvador-Tormo; Manuel Guzmán; Juan Manuel Sepúlveda; Pilar Sánchez-Gómez; Mar Lorente; Guillermo Velasco

Graphical abstract Figure. No Caption available. ABSTRACT Glioblastoma multiforme (GBM) is the most frequent and aggressive type of brain tumor due, at least in part, to its poor response to current anticancer treatments. These features could be explained, at least partially, by the presence within the tumor mass of a small population of cells termed Glioma Initiating Cells (GICs) that has been proposed to be responsible for the relapses occurring in this disease. Thus, the development of novel therapeutic approaches (and specifically those targeting the population of GICs) is urgently needed to improve the survival of the patients suffering this devastating disease. Previous observations by our group and others have shown that &Dgr;9‐Tetrahydrocannabinol (THC, the main active ingredient of marijuana) and other cannabinoids including cannabidiol (CBD) exert antitumoral actions in several animal models of cancer, including gliomas. We also found that the administration of THC (or of THC + CBD at a 1:1 ratio) in combination with temozolomide (TMZ), the benchmark agent for the treatment of GBM, synergistically reduces the growth of glioma xenografts. In this work we investigated the effect of the combination of TMZ and THC:CBD mixtures containing different ratios of the two cannabinoids in preclinical glioma models, including those derived from GICs. Our findings show that TMZ + THC:CBD combinations containing a higher proportion of CDB (but not TMZ + CBD alone) produce a similar antitumoral effect as the administration of TMZ together with THC and CBD at a 1:1 ratio in xenografts generated with glioma cell lines. In addition, we also found that the administration of TMZ + THC:CBD at a 1:1 ratio reduced the growth of orthotopic xenografts generated with GICs derived from GBM patients and enhanced the survival of the animals bearing these intracranial xenografts. Remarkably, the antitumoral effect observed in GICs‐derived xenografts was stronger when TMZ was administered together with cannabinoid combinations containing a higher proportion of CBD. These findings support the notion that the administration of TMZ together with THC:CBD combinations – and specifically those containing a higher proportion of CBD – may be therapeutically explored to target the population of GICs in GBM.


Biochemical Pharmacology | 2018

Optimization of a preclinical therapy of cannabinoids in combination with temozolomide against glioma

Israel López-Valero; Sofia Torres; María Salazar-Roa; Elena García-Taboada; Sonia Hernández-Tiedra; Manuel Guzmán; Juan Manuel Sepúlveda; Guillermo Velasco; Mar Lorente

Graphical abstract Figure. No Caption available. ABSTRACT Glioblastoma multiforme (GBM) is the most frequent and aggressive form of brain cancer. These features are explained at least in part by the high resistance exhibited by these tumors to current anticancer therapies. Thus, the development of novel therapeutic approaches is urgently needed to improve the survival of the patients suffering this devastating disease. &Dgr;9‐Tetrahydrocannabinol (THC, the major active ingredient of marijuana), and other cannabinoids have been shown to exert antitumoral actions in animal models of cancer, including glioma. The mechanism of these anticancer actions relies, at least in part, on the ability of these compounds to stimulate autophagy‐mediated apoptosis in tumor cells. Previous observations from our group demonstrated that local administration of THC (or of THC + CBD at a 1:1 ratio, a mixture that resembles the composition of the cannabinoid‐based medicine Sativex®) in combination with Temozolomide, the benchmark agent for the treatment of GBM, synergistically reduces the growth of glioma xenografts. With the aim of optimizing the possible clinical utilization of cannabinoids in anti‐GBM therapies, in this work we explored the anticancer efficacy of the systemic administration of cannabinoids in combination with TMZ in preclinical models of glioma. Our results show that oral administration of Sativex‐like extracts (containing THC and CBD at a 1:1 ratio) in combination with TMZ produces a strong antitumoral effect in both subcutaneous and intracranial glioma cell‐derived tumor xenografts. In contrast, combined administration of Sativex‐like and BCNU (another alkylating agent used for the treatment of GBM which share structural similarities with the TMZ) did not show a stronger effect than individual treatments. Altogether, our findings support the notion that the combined administration of TMZ and oral cannabinoids could be therapeutically exploited for the management of GBM.

Collaboration


Dive into the Sofia Torres's collaboration.

Top Co-Authors

Avatar

Mar Lorente

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manuel Guzmán

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Sonia Hernández-Tiedra

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

María Salazar

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Elena García-Taboada

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arkaitz Carracedo

University of the Basque Country

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge