Network


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

Hotspot


Dive into the research topics where Saray Garasa is active.

Publication


Featured researches published by Saray Garasa.


Clinical Cancer Research | 2016

Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs)

Carlos Alfaro; Alvaro Teijeira; Carmen Oñate; Guiomar Perez; Miguel F. Sanmamed; Maria Pilar Andueza; Diego Alignani; Sara Labiano; Arantza Azpilikueta; Alfonso Rodriguez-Paulete; Saray Garasa; Juan Pablo Fusco; María Ángela Aznar; Susana Inoges; José Medina-Echeverz; Pedro Berraondo; Jose Luis Perez-Gracia; Ignacio Melero

Purpose: Myeloid-derived suppressor cells (MDSC) are considered an important T-cell immunosuppressive component in cancer-bearing hosts. The factors that attract these cells to the tumor microenvironment are poorly understood. IL8 (CXCL8) is a potent chemotactic factor for neutrophils and monocytes. Experimental Design: MDSC were characterized and sorted by multicolor flow cytometry on ficoll-gradient isolated blood leucokytes from healthy volunteers (n = 10) and advanced cancer patients (n = 28). In chemotaxis assays, sorted granulocytic and monocytic MDSC were tested in response to recombinant IL8, IL8 derived from cancer cell lines, and patient sera. Neutrophil extracellular traps (NETs) formation was assessed by confocal microscopy, fluorimetry, and time-lapse fluorescence confocal microscopy on short-term MDSC cultures. Results: IL8 chemoattracts both granulocytic (GrMDSC) and monocytic (MoMDSC) human MDSC. Monocytic but not granulocytic MDSC exerted a suppressor activity on the proliferation of autologous T cells isolated from the circulation of cancer patients. IL8 did not modify the T-cell suppressor activity of human MDSC. However, IL8 induced the formation of NETs in the GrMDSC subset. Conclusions: IL8 derived from tumors contributes to the chemotactic recruitment of MDSC and to their functional control. Clin Cancer Res; 22(15); 3924–36. ©2016 AACR.


European Journal of Immunology | 2010

Dendritic cells adhere to and transmigrate across lymphatic endothelium in response to IFN‐α

Ana Rouzaut; Saray Garasa; Alvaro Teijeira; Iranzu González; Ivan Martinez-Forero; Natalia Suarez; Esther Larrea; Carlos Alfaro; Asis Palazon; Juan Dubrot; Sandra Hervas-Stubbs; Ignacio Melero

Migration of DC into lymphatic vessels ferries antigenic cargo and pro‐inflammatory stimuli into the draining LN. Given that tissues under the influence of viral infections produce type I IFN, it is conceivable that these cytokines enhance DC migration in order to facilitate an antiviral immune response. Cultured lymphatic endothelium monolayers pretreated with TNF‐α were used to model this phenomenon under inflammatory conditions. DC differentiated in the presence of either IFN‐α2b or IFN‐α5 showed enhanced adhesion to cultured lymphatic endothelial cells. These pro‐adhesive effects were mediated by DC, not the lymphatic endothelium, and correlated with increased DC transmigration across lymphatic endothelial cell monolayers. Transmigration was guided by chemokines acting on DC, and blocking experiments with mAb indicated a role for LFA‐1. Furthermore, incubation of DC with IFN‐α led to the appearance of active conformation epitopes on the CD11a integrin chains expressed by DC. Differentiation of mouse DC in the presence of IFN‐α also increased DC migration from inflammed footpads toward popliteal LN. Collectively, these results indicate a role for type I IFN in directing DC toward LN under inflammatory conditions.


The FASEB Journal | 2012

CD137 on inflamed lymphatic endothelial cells enhances CCL21-guided migration of dendritic cells

Alvaro Teijeira; Asis Palazon; Saray Garasa; Diego Marre; Cristina Aubá; Anne Rogel; Ohiana Murillo; Ivan Martinez-Forero; François Lang; Ignacio Melero; Ana Rouzaut

CD137/TNFR9/41BB was originally described as a surface molecule present on activated T and NK cells. However, its expression is broader among leukocytes, and it is also detected on hypoxic endothelial cells and inflamed blood vessels, as well as in atherosclerotic lesions. Here, we demonstrate that lymphatic endothelial cells (LECs) up‐regulate CD137 expression from undetectable baseline levels on stimulation with TNF‐α, LPS, and IL‐1β. CD137 cross‐linking with an agonistic mAb results in NF‐κB nuclear translocation, followed by up‐regulation of VCAM and a 3‐fold increase in the production of the chemokine CCL21. Accordingly, there is a 50% increase in CCR7‐dependent migration toward conditioned medium from activated LECs on CD137 cross‐linking with the agonistic mAb or the natural ligand (CD137L). Such an enhancement of cell migration is also observed with monocyte‐derived dendritic cells transmigrating across CD137‐activated LEC monolayers. Using explanted human dermal tissue, we found that inflamed skin contains abundant CD137+ lymphatic vessels and that ex vivo incubation of explanted human dermis with TNF‐α induces CD137 expression in lymphatic capillaries. More interestingly, treatment with CD137 agonistic antibody induces CCL21 expression and DC accumulation close to lymphatic vessels. Collectively, our results demonstrate that the inflammatory function of lymphatic vessels can be regulated by CD137.—Teijeira, A., Palazón, A., Garasa, S., Marré, D., Aubá, C., Rogel, A., Murillo, O., Martínez‐Forero, I., Lang, F., Melero, I., Rouzaut, A. CD137 on inflamed lymphatic endothelial cells enhances CCL21‐guided migration of dendritic cells. FASEB J. 26, 3380–3392 (2012). www.fasebj.org


Cancer Research | 2016

Abscopal Effects of Radiotherapy Are Enhanced by Combined Immunostimulatory mAbs and Are Dependent on CD8 T Cells and Crosspriming

Maria E. Rodriguez-Ruiz; Inmaculada Rodriguez; Saray Garasa; Benigno Barbés; Jose Luis Solorzano; Jose Luis Perez-Gracia; Sara Labiano; Miguel F. Sanmamed; Arantza Azpilikueta; Elixabet Bolaños; Alfonso R. Sánchez-Paulete; M. Angela Aznar; Ana Rouzaut; Kurt A. Schalper; Maria Jure-Kunkel; Ignacio Melero

Preclinical and clinical evidence indicate that the proimmune effects of radiotherapy can be synergistically augmented with immunostimulatory mAbs to act both on irradiated tumor lesions and on distant, nonirradiated tumor sites. The combination of radiotherapy with immunostimulatory anti-PD1 and anti-CD137 mAbs was conducive to favorable effects on distant nonirradiated tumor lesions as observed in transplanted MC38 (colorectal cancer), B16OVA (melanoma), and 4T1 (breast cancer) models. The therapeutic activity was crucially performed by CD8 T cells, as found in selective depletion experiments. Moreover, the integrities of BATF-3-dependent dendritic cells specialized in crosspresentation/crosspriming of antigens to CD8+ T cells and of the type I IFN system were absolute requirements for the antitumor effects to occur. The irradiation regimen induced immune infiltrate changes in the irradiated and nonirradiated lesions featured by reductions in the total content of effector T cells, Tregs, and myeloid-derived suppressor cells, while effector T cells expressed more intracellular IFNγ in both the irradiated and contralateral tumors. Importantly, 48 hours after irradiation, CD8+ TILs showed brighter expression of CD137 and PD1, thereby displaying more target molecules for the corresponding mAbs. Likewise, PD1 and CD137 were induced on tumor-infiltrating lymphocytes from surgically excised human carcinomas that were irradiated ex vivo These mechanisms involving crosspriming and CD8 T cells advocate clinical development of immunotherapy combinations with anti-PD1 plus anti-CD137 mAbs that can be synergistically accompanied by radiotherapy strategies, even if the disease is left outside the field of irradiation. Cancer Res; 76(20); 5994-6005. ©2016 AACR.


Journal of Investigative Dermatology | 2013

Lymphatic endothelium forms integrin-engaging 3D structures during DC transit across inflamed lymphatic vessels.

Alvaro Teijeira; Saray Garasa; Rafael Peláez; Arantza Azpilikueta; Carmen Ochoa; Diego Marre; Magda Rodrigues; Carlos Alfaro; Cristina Aubá; Salvatore Valitutti; Ignacio Melero; Ana Rouzaut

Dendritic cell (DC) transmigration across the lymphatic endothelium is critical for the initiation and sustenance of immune responses. Under noninflammatory conditions, DC transit across the lymphatic endothelial cell (LEC) has been shown to be integrin independent. In contrast, there is increasing evidence for the participation of integrins and their ligands in DC transit across lymphatic endothelium under inflammation. In this sense, we describe the formation of ICAM-1 (CD54)-enriched three-dimensional structures on LEC/DC contacts, as these DCs adhere to inflamed skin lymphatic vessels and transmigrate into them. In vitro imaging revealed that under inflammation ICAM-1 accumulated on microvilli projections surrounding 60% of adhered DCs. In contrast, these structures were scarcely formed in noninflammatory conditions. Furthermore, ICAM-1-enriched microvilli were important in promoting DC transendothelial migration and DC crawling over the LEC surface. Microvilli formation was dependent on the presence of β-integrins on the DC side and on integrin conformational affinity to ligand. Finally, we observed that LEC microvilli structures appeared in close vicinity of CCL21 depots and that their assembly was partially inhibited by CCL21-neutralizing antibodies. Therefore, under inflammatory conditions, integrin ligands form three-dimensional membrane projections around DCs. These structures offer docking sites for DC transit from the tissue toward the lymphatic vessel lumen.


Immunology and Cell Biology | 2017

Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells

Maria C. Ochoa; Luna Minute; Inmaculada Rodriguez; Saray Garasa; Elisabeth Perez-Ruiz; Susana Inoges; Ignacio Melero; Pedro Berraondo

Antibody‐dependent cellular cytotoxicity (ADCC) is a set of mechanisms that target cells coated with IgG antibodies of the proper subclasses (IgG1 in the human) to be the prey of cell‐to‐cell cytolysis executed by immune cells expressing FcRIIIA (CD16A). These effectors include not only natural killer (NK) cells but also other CD16+ subsets such as monocyte/macrophages, NKT cells or γδ T cells. In cancer therapy, ADCC is exploited by antibodies that selectively recognize proteins on the surface of malignant cells. An approach to enhance antitumor activity is to act on effector cells so they are increased in their numbers or enhanced in their individual (on a cell per cell basis) ADCC performance. This enhancement can be therapeutically attained by cytokines (that is, interleukin (IL)‐15, IL‐21, IL‐18, IL‐2); immunostimulatory monoclonal antibodies (that is, anti‐CD137, anti‐CD96, anti‐TIGIT, anti‐KIR, anti‐PD‐1); TLR agonists or by adoptive infusions of ex vivo expanded NK cells which can be genetically engineered to become more efficient effectors. In conjunction with approaches optimizing IgG1 Fc affinity to CD16, acting on effector cells offers hope to achieve synergistic immunotherapy strategies.


Molecular Cancer | 2014

Combined targeting of TGF-β1 and integrin β3 impairs lymph node metastasis in a mouse model of non-small-cell lung cancer

Elizabeth Salvo; Saray Garasa; Javier Dotor; Xabier Morales; Rafael Peláez; Peter Altevogt; Ana Rouzaut

BackgroundTransforming Growth Factor beta (TGF-β) acts as a tumor suppressor early in carcinogenesis but turns into tumor promoter in later disease stages. In fact, TGF-β is a known inducer of integrin expression by tumor cells which contributes to cancer metastatic spread and TGF-β inhibition has been shown to attenuate metastasis in mouse models. However, carcinoma cells often become refractory to TGF-β-mediated growth inhibition. Therefore identifying patients that may benefit from anti-TGF-β therapy requires careful selection.MethodsWe performed in vitro analysis of the effects of exposure to TGF-β in NSCLC cell chemotaxis and adhesion to lymphatic endothelial cells. We also studied in an orthotopic model of NSCLC the incidence of metastases to the lymph nodes after inhibition of TGF-β signaling, β3 integrin expression or both.ResultsWe offer evidences of increased β3-integrin dependent NSCLC adhesion to lymphatic endothelium after TGF-β exposure. In vivo experiments show that targeting of TGF-β and β3 integrin significantly reduces the incidence of lymph node metastasis. Even more, blockade of β3 integrin expression in tumors that did not respond to TGF-β inhibition severely impaired the ability of the tumor to metastasize towards the lymph nodes.ConclusionThese findings suggest that lung cancer tumors refractory to TGF-β monotherapy can be effectively treated using dual therapy that combines the inhibition of tumor cell adhesion to lymphatic vessels with stromal TGF-β inhibition.


International Journal of Cancer | 2013

Phosphorylated tubulin adaptor protein CRMP-2 as prognostic marker and candidate therapeutic target for NSCLC.

Erik Oliemuller; Rafael Peláez; Saray Garasa; Maria J. Pajares; Jackeline Agorreta; Ruben Pio; Luis M. Montuenga; Alvaro Teijeira; Susana Llanos; Ana Rouzaut

Collapsin response mediator protein‐2 (CRMP‐2) is the first described and most studied member of a family of proteins that mediate the addition of tubulin dimers to the growing microtubule. CRMPs have mainly been studied in the nervous system, but recently, they have been described in other tissues where they participate in vesicle transport, migration and mitosis. In this work, we aimed at studying the role of CRMP‐2 in lung cancer cell division. We first explored the expression of CRMP‐2 and phosphorylated (Thr 514) CRMP‐2 in 91 samples obtained from patients with localized nonsmall cell lung cancer. We observed a significant correlation between high levels of nuclear phosphorylated CRMP‐2 and poor prognosis in those patients. Interestingly, this association was only positive for untreated patients. To provide a mechanistic explanation to these findings, we used in vitro models to analyze the role of CRMP‐2 and its phosphorylated forms in cell division. Thus, we observed by confocal microscopy and immunoprecipitation assays that CRMP‐2 differentially colocalizes with the mitotic spindle during cell division. The use of phosphodefective or phosphomimetic mutants of CRMP‐2 allowed us to prove that anomalies in the phosphorylation status of CRMP‐2 result in changes in the mitotic tempo, and increments in the number of multinucleated cells. Finally, here we demonstrate that CRMP‐2 phosphorylation impairment, or silencing induces p53 expression and promotes apoptosis through caspase 3 activation. These results pointed to CRMP‐2 phosphorylation as a prognostic marker and potential new target to be explored in cancer therapy.


International Journal of Radiation Oncology Biology Physics | 2017

Intercellular Adhesion Molecule-1 and Vascular Cell Adhesion Molecule Are Induced by Ionizing Radiation on Lymphatic Endothelium

Maria E. Rodriguez-Ruiz; Saray Garasa; Inmaculada Rodriguez; Jose Luis Solorzano; Benigno Barbés; Alba Yanguas; Alvaro Teijeira; Iñaki Etxeberria; J. Aristu; Cornelia Halin; Ignacio Melero; Ana Rouzaut

PURPOSE/OBJECTIVES The goal of this study was to assess the effects of ionizing radiation on the expression of the integrin ligands ICAM-1 and VCAM that control leucocyte transit by lymphatic endothelial cells. MATERIALS/METHODS Confluent monolayers of primary human lymphatic endothelial cells (LEC) were irradiated with single dose of 2, 5, 10 or 20 Gy, with 6 MeV-x-rays using a Linear-Accelerator. ICAM-1 and VCAM expression was determined by flow cytometry. Human tissue specimens received a single dose of 20 Gy with 15 MeV-x-rays. MC38, B16-OVA or B16-VEGF-C tumors grown in C57BL/6 mice were irradiated with single dose of 20Gy using a Linear-Accelerator fitted with a 10mm Radiosurgery collimator. Clinical samples were obtained from patients previous and 4 weeks after complete standard radiotherapy. ICAM-1 and VCAM expression was detected in all tissue specimens by confocal microscopy. To understand the role of TGFβ in this process anti-TGFβ blocking mAb were injected i.p. 30min before radiotherapy. Cell adhesion to irradiated LEC was analyzed in adhesion experiments performed in the presence or in the absence of anti- TGFβ and /or anti-ICAM1 blocking mAb. RESULTS We demonstrate that lymphatic endothelial cells in tumor samples experience induction of surface ICAM-1 and VCAM when exposed to ionizing radiation in a dose- and time-dependent manner. These effects can be recapitulated in cultured LEC, and are in part mediated by TGFβ. These data are consistent with increases in ICAM-1 and VCAM expression on LYVE-1+ endothelial cells in freshly explanted human tumor tissue and in mouse transplanted tumors after radiotherapy. Finally, ICAM-1 and VCAM expression accounts for enhanced adherence of human T lymphocytes to irradiated LEC. CONCLUSION Our results show induction of ICAM-1 and VCAM on LVs in irradiated lesions and offer a starting point for elucidating the biological and therapeutic implications of targeting leukocyte traffic in combination to immunotherapy.


Annals of Oncology | 2017

Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy

Alfonso R. Sánchez-Paulete; Alvaro Teijeira; Francisco J. Cueto; Saray Garasa; Jose Luis Perez-Gracia; A Sánchez-Arráez; David Sancho; Ignacio Melero

Dendritic cells (DCs) are the main professional antigen-presenting cells for induction of T-cell adaptive responses. Cancer cells express tumor antigens, including neoantigens generated by nonsynonymous mutations, but are poor for antigen presentation and for providing costimulatory signals for T-cell priming. Mounting evidence suggests that antigen transfer to DCs and their surrogate presentation on major histocompatibility complex class I and II molecules together with costimulatory signals is paramount for induction of viral and cancer immunity. Of the great diversity of DCs, BATF3/IRF8-dependent conventional DCs type 1 (cDC1) excel at cross-presentation of tumor cell-associated antigens. Location of cDC1s in the tumor correlates with improved infiltration by CD8+ T cells and tumor-specific T-cell immunity. Indeed, cDC1s are crucial for antitumor efficacy using checkpoint inhibitors and anti-CD137 agonist monoclonal antibodies in mouse models. Enhancement and exploitation of T-cell cross-priming by cDC1s offer opportunities for improved cancer immunotherapy, including in vivo targeting of tumor antigens to internalizing receptors on cDC1s and strategies to increase their numbers, activation and priming capacity within tumors and tumor-draining lymph nodes.

Collaboration


Dive into the Saray Garasa'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