93 results on '"Uranga Murillo, Iratxe"'
Search Results
2. Identification of FasL as a crucial host factor driving COVID-19 pathology and lethality
- Author
-
Albert, Marie-Christine, Uranga-Murillo, Iratxe, Arias, Maykel, De Miguel, Diego, Peña, Natacha, Montinaro, Antonella, Varanda, Ana Beatriz, Theobald, Sebastian J., Areso, Itziar, Saggau, Julia, Koch, Manuel, Liccardi, Gianmaria, Peltzer, Nieves, Rybniker, Jan, Hurtado-Guerrero, Ramón, Merino, Pedro, Monzón, Marta, Badiola, Juan J., Reindl-Schwaighofer, Roman, Sanz-Pamplona, Rebeca, Cebollada-Solanas, Alberto, Megyesfalvi, Zsolt, Dome, Balazs, Secrier, Maria, Hartmann, Boris, Bergmann, Michael, Pardo, Julián, and Walczak, Henning
- Published
- 2024
- Full Text
- View/download PDF
3. Intravenous administration of BCG in mice promotes natural killer and T cell-mediated antitumor immunity in the lung
- Author
-
Moreo, Eduardo, Jarit-Cabanillas, Aitor, Robles-Vera, Iñaki, Uranga, Santiago, Guerrero, Claudia, Gómez, Ana Belén, Mata-Martínez, Pablo, Minute, Luna, Araujo-Voces, Miguel, Felgueres, María José, Esteso, Gloria, Uranga-Murillo, Iratxe, Arias, Maykel, Pardo, Julián, Martín, Carlos, Valés-Gómez, Mar, del Fresno, Carlos, Sancho, David, and Aguiló, Nacho
- Published
- 2023
- Full Text
- View/download PDF
4. Ultrastructural analysis and three-dimensional reconstruction of cellular structures involved in SARS-CoV-2 spread
- Author
-
Baselga, Marta, Moreo, Eduardo, Uranga-Murillo, Iratxe, Arias, Maykel, and Junquera, Concepción
- Published
- 2023
- Full Text
- View/download PDF
5. Immune response in SARS-CoV-2 infection
- Author
-
Morte Romea, Elena [0000-0001-9262-2461], Uranga Murillo, Iratxe [0000-0001-8411-984X], Hidalgo, Sandra [0000-0003-1629-9978], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Miguel, Diego de [0000-0002-8486-8514], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Martínez Lostao, Luis [0000-0003-3043-147X], Pellejero, Galadriel [0000-0002-2728-5435], Pardo, Julián [0000-0003-0154-0730], Paño, José Ramón [0000-0002-9600-8116], Morte Romea, Elena, Uranga Murillo, Iratxe, Hidalgo, Sandra, Ramírez-Labrada, Ariel, Miguel, Diego de, Gálvez Buerba, Eva Mª, Martínez Lostao, Luis, Pellejero, Galadriel, Letona Jiménez, Santiago, Pardo, Julián, Paño, José Ramón, Morte Romea, Elena [0000-0001-9262-2461], Uranga Murillo, Iratxe [0000-0001-8411-984X], Hidalgo, Sandra [0000-0003-1629-9978], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Miguel, Diego de [0000-0002-8486-8514], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Martínez Lostao, Luis [0000-0003-3043-147X], Pellejero, Galadriel [0000-0002-2728-5435], Pardo, Julián [0000-0003-0154-0730], Paño, José Ramón [0000-0002-9600-8116], Morte Romea, Elena, Uranga Murillo, Iratxe, Hidalgo, Sandra, Ramírez-Labrada, Ariel, Miguel, Diego de, Gálvez Buerba, Eva Mª, Martínez Lostao, Luis, Pellejero, Galadriel, Letona Jiménez, Santiago, Pardo, Julián, and Paño, José Ramón
- Abstract
We analyzed several parameters of the immune response of COVID-19 patients who required hospital admission, com paring them to healthy donors (HDs) and other patients admitted for respiratory infections not caused by SARS-CoV-2 (non-COV-RTI). We focused on the main cells involved in innate immunity and soluble inflammatory factors that regulate their activity.
- Published
- 2024
6. Identification of FasL as a crucial host factor driving COVID-19 pathology and lethality
- Author
-
Albert, Marie Christine, Uranga-Murillo, Iratxe, Arias, Maykel, De Miguel, Diego, Peña, Natacha, Montinaro, Antonella, Varanda, Ana Beatriz, Theobald, Sebastian J., Areso, Itziar, Saggau, Julia, Koch, Manuel, Liccardi, Gianmaria, Peltzer, Nieves, Rybniker, Jan, Hurtado-Guerrero, Ramón, Merino, Pedro, Monzón, Marta, Badiola, Juan J., Reindl-Schwaighofer, Roman, Sanz-Pamplona, Rebeca, Cebollada-Solanas, Alberto, Megyesfalvi, Zsolt, Dome, Balazs, Secrier, Maria, Hartmann, Boris, Bergmann, Michael, Pardo, Julián, Walczak, Henning, Albert, Marie Christine, Uranga-Murillo, Iratxe, Arias, Maykel, De Miguel, Diego, Peña, Natacha, Montinaro, Antonella, Varanda, Ana Beatriz, Theobald, Sebastian J., Areso, Itziar, Saggau, Julia, Koch, Manuel, Liccardi, Gianmaria, Peltzer, Nieves, Rybniker, Jan, Hurtado-Guerrero, Ramón, Merino, Pedro, Monzón, Marta, Badiola, Juan J., Reindl-Schwaighofer, Roman, Sanz-Pamplona, Rebeca, Cebollada-Solanas, Alberto, Megyesfalvi, Zsolt, Dome, Balazs, Secrier, Maria, Hartmann, Boris, Bergmann, Michael, Pardo, Julián, and Walczak, Henning
- Abstract
The dysregulated immune response and inflammation resulting in severe COVID-19 are still incompletely understood. Having recently determined that aberrant death-ligand-induced cell death can cause lethal inflammation, we hypothesized that this process might also cause or contribute to inflammatory disease and lung failure following SARS-CoV-2 infection. To test this hypothesis, we developed a novel mouse-adapted SARS-CoV-2 model (MA20) that recapitulates key pathological features of COVID-19. Concomitantly with occurrence of cell death and inflammation, FasL expression was significantly increased on inflammatory monocytic macrophages and NK cells in the lungs of MA20-infected mice. Importantly, therapeutic FasL inhibition markedly increased survival of both, young and old MA20-infected mice coincident with substantially reduced cell death and inflammation in their lungs. Intriguingly, FasL was also increased in the bronchoalveolar lavage fluid of critically-ill COVID-19 patients. Together, these results identify FasL as a crucial host factor driving the immuno-pathology that underlies COVID-19 severity and lethality, and imply that patients with severe COVID-19 may significantly benefit from therapeutic inhibition of FasL., The dysregulated immune response and inflammation resulting in severe COVID-19 are still incompletely understood. Having recently determined that aberrant death-ligand-induced cell death can cause lethal inflammation, we hypothesized that this process might also cause or contribute to inflammatory disease and lung failure following SARS-CoV-2 infection. To test this hypothesis, we developed a novel mouse-adapted SARS-CoV-2 model (MA20) that recapitulates key pathological features of COVID-19. Concomitantly with occurrence of cell death and inflammation, FasL expression was significantly increased on inflammatory monocytic macrophages and NK cells in the lungs of MA20-infected mice. Importantly, therapeutic FasL inhibition markedly increased survival of both, young and old MA20-infected mice coincident with substantially reduced cell death and inflammation in their lungs. Intriguingly, FasL was also increased in the bronchoalveolar lavage fluid of critically-ill COVID-19 patients. Together, these results identify FasL as a crucial host factor driving the immuno-pathology that underlies COVID-19 severity and lethality, and imply that patients with severe COVID-19 may significantly benefit from therapeutic inhibition of FasL.
- Published
- 2024
7. Supplementary material for Integrated analysis of circulating immune cellular and soluble mediators reveals specific COVID19 signatures at hospital admission with utility for prediction of clinical outcomes [Dataset]
- Author
-
European Commission, Gobierno de Aragón, Instituto de Salud Carlos III, Fundación Banco Santander, Universidad de Zaragoza, Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, Asociación Española Contra el Cáncer, Ministerio de Ciencia, Innovación y Universidades (España), ARAID Foundation, Uranga Murillo, Iratxe [0000-0001-8411-984X], Morte Romea, Elena [0000-0001-9262-2461], Hidalgo, Sandra [0000-0003-1629-9978], Pesini, Cecilia [0000-0002-8707-2722], García-Mulero, Sandra [0000-0003-4931-1267], Sierra Monzón, José L. [0000-0002-8796-2717], Santiago, Llipsy [0000-0002-1861-5981], Arias, Maykel [0000-0002-9730-2210], Miguel, Diego de [0000-0002-8486-8514], Encabo-Berzosa, M. Mar [0000-0001-5533-804X], Gracia Tello, Borja [0000-0003-3248-2908], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], Martínez Lostao, Luis [0000-0003-3043-147X], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Paño, José Ramón [0000-0002-9600-8116], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Pardo, Julián [0000-0003-0154-0730], Paño, José Ramón; Ramírez-Labrada, Ariel; Pardo, Julián, Uranga Murillo, Iratxe, Morte Romea, Elena, Hidalgo, Sandra, Pesini, Cecilia, García-Mulero, Sandra, Sierra Monzón, José L., Santiago, Llipsy, Arias, Maykel, Miguel, Diego de, Encabo-Berzosa, M. Mar, Gracia Tello, Borja, Sanz-Pamplona, Rebeca, Martínez-Lostao, Luis, Gálvez Buerba, Eva Mª, Paño, José Ramón, Ramírez-Labrada, Ariel, Pardo, Julián, European Commission, Gobierno de Aragón, Instituto de Salud Carlos III, Fundación Banco Santander, Universidad de Zaragoza, Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, Asociación Española Contra el Cáncer, Ministerio de Ciencia, Innovación y Universidades (España), ARAID Foundation, Uranga Murillo, Iratxe [0000-0001-8411-984X], Morte Romea, Elena [0000-0001-9262-2461], Hidalgo, Sandra [0000-0003-1629-9978], Pesini, Cecilia [0000-0002-8707-2722], García-Mulero, Sandra [0000-0003-4931-1267], Sierra Monzón, José L. [0000-0002-8796-2717], Santiago, Llipsy [0000-0002-1861-5981], Arias, Maykel [0000-0002-9730-2210], Miguel, Diego de [0000-0002-8486-8514], Encabo-Berzosa, M. Mar [0000-0001-5533-804X], Gracia Tello, Borja [0000-0003-3248-2908], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], Martínez Lostao, Luis [0000-0003-3043-147X], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Paño, José Ramón [0000-0002-9600-8116], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Pardo, Julián [0000-0003-0154-0730], Paño, José Ramón; Ramírez-Labrada, Ariel; Pardo, Julián, Uranga Murillo, Iratxe, Morte Romea, Elena, Hidalgo, Sandra, Pesini, Cecilia, García-Mulero, Sandra, Sierra Monzón, José L., Santiago, Llipsy, Arias, Maykel, Miguel, Diego de, Encabo-Berzosa, M. Mar, Gracia Tello, Borja, Sanz-Pamplona, Rebeca, Martínez-Lostao, Luis, Gálvez Buerba, Eva Mª, Paño, José Ramón, Ramírez-Labrada, Ariel, and Pardo, Julián
- Abstract
Supplementary materials and methods: sample processing, flow cytometry, high dimensional flow cytometry data analysis, multiplex plasma protein analyses, granzyme activity assay in serum, statistics. Supplementary figure legends (1-5). Supplementary table legends (1-7).
- Published
- 2022
8. All about (NK cell-mediated) death in two acts and an unexpected encore: initiation, execution and activation of adaptive immunity
- Author
-
Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Gobierno de Aragón, Agencia Estatal de Investigación (España), Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, Asociación Española Contra el Cáncer, Ministerio de Ciencia, Innovación y Universidades (España), Ramírez-Labrada, Ariel [0000-0002-3888-7036], Pesini, Cecilia [0000-0002-8707-2722], Santiago, Llipsy [0000-0002-1861-5981], Hidalgo, Sandra [0000-0003-1629-9978], Uranga Murillo, Iratxe [0000-0001-8411-984X], Arias, Maykel [0000-0002-9730-2210], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Pardo, Julián [0000-0003-0154-0730], Ramírez-Labrada, Ariel, Pesini, Cecilia, Santiago, Llipsy, Hidalgo, Sandra, Calvo Pérez, Adanys, Oñate, Carmen, Andrés Tovar, Alejandro, Garzón Tituaña, Marcela, Uranga Murillo, Iratxe, Arias, Maykel, Gálvez Buerba, Eva Mª, Pardo, Julián, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Gobierno de Aragón, Agencia Estatal de Investigación (España), Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, Asociación Española Contra el Cáncer, Ministerio de Ciencia, Innovación y Universidades (España), Ramírez-Labrada, Ariel [0000-0002-3888-7036], Pesini, Cecilia [0000-0002-8707-2722], Santiago, Llipsy [0000-0002-1861-5981], Hidalgo, Sandra [0000-0003-1629-9978], Uranga Murillo, Iratxe [0000-0001-8411-984X], Arias, Maykel [0000-0002-9730-2210], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Pardo, Julián [0000-0003-0154-0730], Ramírez-Labrada, Ariel, Pesini, Cecilia, Santiago, Llipsy, Hidalgo, Sandra, Calvo Pérez, Adanys, Oñate, Carmen, Andrés Tovar, Alejandro, Garzón Tituaña, Marcela, Uranga Murillo, Iratxe, Arias, Maykel, Gálvez Buerba, Eva Mª, and Pardo, Julián
- Abstract
NK cells are key mediators of immune cell-mediated cytotoxicity toward infected and transformed cells, being one of the main executors of cell death in the immune system. NK cells recognize target cells through an array of inhibitory and activating receptors for endogenous or exogenous pathogen-derived ligands, which together with adhesion molecules form a structure known as immunological synapse that regulates NK cell effector functions. The main and best characterized mechanisms involved in NK cell-mediated cytotoxicity are the granule exocytosis pathway (perforin/granzymes) and the expression of death ligands. These pathways are recognized as activators of different cell death programmes on the target cells leading to their destruction. However, most studies analyzing these pathways have used pure recombinant or native proteins instead of intact NK cells and, thus, extrapolation of the results to NK cell-mediated cell death might be difficult. Specially, since the activation of granule exocytosis and/or death ligands during NK cell-mediated elimination of target cells might be influenced by the stimulus received from target cells and other microenvironment components, which might affect the cell death pathways activated on target cells. Here we will review and discuss the available experimental evidence on how NK cells kill target cells, with a special focus on the different cell death modalities that have been found to be activated during NK cell-mediated cytotoxicity; including apoptosis and more inflammatory pathways like necroptosis and pyroptosis. In light of this new evidence, we will develop the new concept of cell death induced by NK cells as a new regulatory mechanism linking innate immune response with the activation of tumour adaptive T cell responses, which might be the initiating stimulus that trigger the cancer-immunity cycle. The use of the different cell death pathways and the modulation of the tumour cell molecular machinery regulating them might
- Published
- 2022
9. Adoptive NK cell transfer as a treatment in colorectal cancer patients: analyses of tumour cell determinants correlating with efficacy in vitro and in vivo
- Author
-
Aspanoa, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Fundación Inocente Inocente, Asociación Carrera de la Mujer Ciudad de Monzón, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), ARAID Foundation, Institut Català d'Oncologia, Ministerio de Economía, Industria y Competitividad (España), Centro de Investigación Biomédica en Red Epidemiología y Salud Pública (España), Agència de Gestió d'Ajuts Universitaris i de Recerca, European Cooperation in Science and Technology, Lanuza, Pilar M. [0000-0001-7328-2094], Alonso, M. Henar [0000-0003-0285-5451], Hidalgo, Sandra [0000-0003-1629-9978], Uranga Murillo, Iratxe [0000-0001-8411-984X], García-Mulero, Sandra [0000-0003-4931-1267], Sanjuan, Xavier [0000-0002-5253-822X], Santiago, Llipsy [0000-0002-1861-5981], Comas, Laura [0000-0002-3843-1231], Redrado, Sergio [0000-0002-8404-0012], Pazo-Cid, Roberto [0000-0002-8026-7391], Jaime Sánchez, Paula [0000-0002-8731-4269], Pesini, Cecilia [0000-0002-8707-2722], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Arias, Maykel [0000-0002-9730-2210], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], Pardo, Julián [0000-0003-0154-0730], Lanuza, Pilar M., Alonso, M. Henar, Hidalgo, Sandra, Uranga Murillo, Iratxe, García-Mulero, Sandra, Arnau, Raquel, Santos Vivas, Cristina, Sanjuan, Xavier, Santiago, Llipsy, Comas, Laura, Redrado, Sergio, Pazo-Cid, Roberto, Agustín Ferrández, M. J., Jaime Sánchez, Paula, Pesini, Cecilia, Gálvez Buerba, Eva Mª, Ramírez-Labrada, Ariel, Arias, Maykel, Sanz-Pamplona, Rebeca, Pardo, Julián, Aspanoa, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Fundación Inocente Inocente, Asociación Carrera de la Mujer Ciudad de Monzón, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), ARAID Foundation, Institut Català d'Oncologia, Ministerio de Economía, Industria y Competitividad (España), Centro de Investigación Biomédica en Red Epidemiología y Salud Pública (España), Agència de Gestió d'Ajuts Universitaris i de Recerca, European Cooperation in Science and Technology, Lanuza, Pilar M. [0000-0001-7328-2094], Alonso, M. Henar [0000-0003-0285-5451], Hidalgo, Sandra [0000-0003-1629-9978], Uranga Murillo, Iratxe [0000-0001-8411-984X], García-Mulero, Sandra [0000-0003-4931-1267], Sanjuan, Xavier [0000-0002-5253-822X], Santiago, Llipsy [0000-0002-1861-5981], Comas, Laura [0000-0002-3843-1231], Redrado, Sergio [0000-0002-8404-0012], Pazo-Cid, Roberto [0000-0002-8026-7391], Jaime Sánchez, Paula [0000-0002-8731-4269], Pesini, Cecilia [0000-0002-8707-2722], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Arias, Maykel [0000-0002-9730-2210], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], Pardo, Julián [0000-0003-0154-0730], Lanuza, Pilar M., Alonso, M. Henar, Hidalgo, Sandra, Uranga Murillo, Iratxe, García-Mulero, Sandra, Arnau, Raquel, Santos Vivas, Cristina, Sanjuan, Xavier, Santiago, Llipsy, Comas, Laura, Redrado, Sergio, Pazo-Cid, Roberto, Agustín Ferrández, M. J., Jaime Sánchez, Paula, Pesini, Cecilia, Gálvez Buerba, Eva Mª, Ramírez-Labrada, Ariel, Arias, Maykel, Sanz-Pamplona, Rebeca, and Pardo, Julián
- Abstract
[Background]: Colorectal cancer (CRC) is a heterogeneous disease with variable mutational profile and tumour microenvironment composition that influence tumour progression and response to treatment. While chemoresistant and poorly immunogenic CRC remains a challenge, the development of new strategies guided by biomarkers could help stratify and treat patients. Allogeneic NK cell transfer emerges as an alternative against chemoresistant and poorly immunogenic CRC., [Methods]: NK cell-related immunological markers were analysed by transcriptomics and immunohistochemistry in human CRC samples and correlated with tumour progression and overall survival. The anti-tumour ability of expanded allogeneic NK cells using a protocol combining cytokines and feeder cells was analysed in vitro and in vivo and correlated with CRC mutational status and the expression of ligands for immune checkpoint (IC) receptors regulating NK cell activity., [Results]: HLA-I downmodulation and NK cell infiltration correlated with better overall survival in patients with a low-stage (II) microsatellite instability-high (MSI-H) CRC, suggesting a role of HLA-I as a prognosis biomarker and a potential benefit of NK cell immunotherapy. Activated allogeneic NK cells were able to eliminate CRC cultures without PD-1 and TIM-3 restriction but were affected by HLA-I expression. In vivo experiments confirmed the efficacy of the therapy against both HLA+ and HLA− CRC cell lines. Concomitant administration of pembrolizumab failed to improve tumour control., [Conclusions]: Our results reveal an immunological profile of CRC tumours in which immunogenicity (MSI-H) and immune evasion mechanisms (HLA downmodulation) favour NK cell immunosurveillance at early disease stages. Accordingly, we have shown that allogeneic NK cell therapy can target tumours expressing mutations conferring poor prognosis regardless of the expression of T cell-related inhibitory IC ligands. Overall, this study provides a rationale for a new potential basis for CRC stratification and NK cell-based therapy.
- Published
- 2022
10. Antigen-specific primed cytotoxic T cells eliminate tumour cells in vivo and prevent tumour development, regardless of the presence of anti-apoptotic mutations conferring drug resistance
- Author
-
Jaime-Sánchez, Paula, Catalán, Elena, Uranga-Murillo, Iratxe, Aguiló, Nacho, Santiago, Llipsy, M Lanuza, Pilar, de Miguel, Diego, A Arias, Maykel, and Pardo, Julián
- Published
- 2018
- Full Text
- View/download PDF
11. Cleavage of cFLIP restrains cell death during viral infection and tissue injury and favors tissue repair
- Author
-
Martinez Lagunas, Kristel, primary, Savcigil, Deniz Pinar, additional, Zrilic, Matea, additional, Carvajal Fraile, Carlos, additional, Craxton, Andrew, additional, Self, Emily, additional, Uranga-Murillo, Iratxe, additional, de Miguel, Diego, additional, Arias, Maykel, additional, Willenborg, Sebastian, additional, Piekarek, Michael, additional, Albert, Marie Christine, additional, Nugraha, Kalvin, additional, Lisewski, Ina, additional, Janakova, Erika, additional, Igual, Natalia, additional, Tonnus, Wulf, additional, Hildebrandt, Ximena, additional, Ibrahim, Mohammed, additional, Ballegeer, Marlies, additional, Saelens, Xavier, additional, Kueh, Andrew, additional, Meier, Pascal, additional, Linkermann, Andreas, additional, Pardo, Julian, additional, Eming, Sabine, additional, Walczak, Henning, additional, MacFarlane, Marion, additional, Peltzer, Nieves, additional, and Annibaldi, Alessandro, additional
- Published
- 2022
- Full Text
- View/download PDF
12. Systemic BCG administration stimulates NK and T cell-mediated antitumor lung immunity and overcomes tumor resistance to immune checkpoint inhibition
- Author
-
AGUILO, NACHO, primary, Moreo, Eduardo, additional, Robles, Iñaqui, additional, Guerrero, Claudia, additional, Jarit, Aitor, additional, Felgueres, María José, additional, Esteso, Gloria, additional, Uranga-Murillo, Iratxe, additional, Uranga, Santiago, additional, Gomez, Ana Belen, additional, Pardo, Julian, additional, Martin, Carlos, additional, Vales-Gomez, Mar, additional, Fresno, Carlos del, additional, and Sancho, David, additional
- Published
- 2022
- Full Text
- View/download PDF
13. Granzyme A inhibition reduces inflammation and increases survival during abdominal sepsis
- Author
-
Garzón, Marcela [0000-0001-6778-0636], Comas, Laura [0000-0002-3843-1231], Santiago, Llipsy [0000-0002-1861-5981], Uranga Murillo, Iratxe [0000-0001-8411-984X], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Pardo, Julián [0000-0003-0154-0730], Arias, Maykel [0000-0002-9730-2210], Garzón, Marcela, Comas, Laura, Santiago, Llipsy, Uranga Murillo, Iratxe, Ramírez-Labrada, Ariel, Gálvez Buerba, Eva Mª, Pardo, Julián, Arias, Maykel, Garzón, Marcela [0000-0001-6778-0636], Comas, Laura [0000-0002-3843-1231], Santiago, Llipsy [0000-0002-1861-5981], Uranga Murillo, Iratxe [0000-0001-8411-984X], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Pardo, Julián [0000-0003-0154-0730], Arias, Maykel [0000-0002-9730-2210], Garzón, Marcela, Comas, Laura, Santiago, Llipsy, Uranga Murillo, Iratxe, Ramírez-Labrada, Ariel, Gálvez Buerba, Eva Mª, Pardo, Julián, and Arias, Maykel
- Abstract
Peritonitis is one of the most common causes of sepsis. Evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed by NK and T cells, could act as proinflammatory mediator and could play an important role in the pathogenesis of sepsis. Here, we aim to analyze the role and therapeutic potential of GzmA in the pathogenesis of peritoneal sepsis.
- Published
- 2021
14. Determination of the concentration of IgG against the spike receptor-binding domain that predicts the viral neutralizing activity of convalescent plasma and serum against SARS-CoV-2
- Author
-
Gobierno de Aragón, Fundación Banco Santander, Universidad de Zaragoza, Instituto de Salud Carlos III, European Commission, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, Santiago, Llipsy [0000-0002-1861-5981], Uranga Murillo, Iratxe [0000-0001-8411-984X], Arias, Maykel [0000-0002-9730-2210], González Ramírez, Andrés Manuel [0000-0002-5838-0857], Macías León, Javier [0000-0001-6815-6720], Moreo, Eduardo [0000-0002-0182-201X], Redrado, Sergio [0000-0002-8404-0012], Taleb, V. [0000-0001-9224-5854], Lira-Navarrete, Erandi [0000-0003-2462-7580], Hurtado-Guerrero, R. [0000-0002-3122-9401], Encabo-Berzosa, M. Mar [0000-0001-5533-804X], Hidalgo, Sandra [0000-0003-1629-9978], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Miguel, Diego de [0000-0002-8486-8514], Benito, Rafael [0000-0001-5134-1006], Fernández Casanovas, Antonio [0000-0002-2557-4890], Serrano Barcos, Laura [0000-0002-9291-9801], Yuste, Cristina [0000-0003-3667-523X], Villanueva-Saz, Sergio [0000-0001-6209-4282], Paño, José Ramón [0000-0002-9600-8116], Pardo, Julián [0000-0003-0154-0730], Santiago, Llipsy, Uranga Murillo, Iratxe, Arias, Maykel, González Ramírez, Andrés Manuel, Macías-León, Javier, Moreo, Eduardo, Redrado, Sergio, García García, Ana, Taleb, V., Lira-Navarrete, Erandi, Hurtado-Guerrero, R., Aguiló, Nacho, Encabo-Berzosa, M. Mar, Hidalgo, Sandra, Gálvez Buerba, Eva Mª, Ramírez-Labrada, Ariel, Miguel, Diego de, Benito, Rafael, Miranda, Patricia, Fernández Casanovas, Antonio, Domingo, José María, Serrano Barcos, Laura, Yuste, Cristina, Villanueva-Saz, Sergio, Paño, José Ramón, Pardo, Julián, Gobierno de Aragón, Fundación Banco Santander, Universidad de Zaragoza, Instituto de Salud Carlos III, European Commission, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, Santiago, Llipsy [0000-0002-1861-5981], Uranga Murillo, Iratxe [0000-0001-8411-984X], Arias, Maykel [0000-0002-9730-2210], González Ramírez, Andrés Manuel [0000-0002-5838-0857], Macías León, Javier [0000-0001-6815-6720], Moreo, Eduardo [0000-0002-0182-201X], Redrado, Sergio [0000-0002-8404-0012], Taleb, V. [0000-0001-9224-5854], Lira-Navarrete, Erandi [0000-0003-2462-7580], Hurtado-Guerrero, R. [0000-0002-3122-9401], Encabo-Berzosa, M. Mar [0000-0001-5533-804X], Hidalgo, Sandra [0000-0003-1629-9978], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Miguel, Diego de [0000-0002-8486-8514], Benito, Rafael [0000-0001-5134-1006], Fernández Casanovas, Antonio [0000-0002-2557-4890], Serrano Barcos, Laura [0000-0002-9291-9801], Yuste, Cristina [0000-0003-3667-523X], Villanueva-Saz, Sergio [0000-0001-6209-4282], Paño, José Ramón [0000-0002-9600-8116], Pardo, Julián [0000-0003-0154-0730], Santiago, Llipsy, Uranga Murillo, Iratxe, Arias, Maykel, González Ramírez, Andrés Manuel, Macías-León, Javier, Moreo, Eduardo, Redrado, Sergio, García García, Ana, Taleb, V., Lira-Navarrete, Erandi, Hurtado-Guerrero, R., Aguiló, Nacho, Encabo-Berzosa, M. Mar, Hidalgo, Sandra, Gálvez Buerba, Eva Mª, Ramírez-Labrada, Ariel, Miguel, Diego de, Benito, Rafael, Miranda, Patricia, Fernández Casanovas, Antonio, Domingo, José María, Serrano Barcos, Laura, Yuste, Cristina, Villanueva-Saz, Sergio, Paño, José Ramón, and Pardo, Julián
- Abstract
Passive immunization with hyperimmune plasma from convalescent patients has been proposed as a potentially useful treatment for COVID-19. Nevertheless, its efficacy in patients with COVID-19 remains uncertain. Thus, the establishment and validation of standardized methods that predict the viral neutralizing (VN) activity of plasma against SARS-CoV-2 is of utmost importance to appraise its therapeutic value. Using an in-house quantitative ELISA test and two independent cohorts with a total of 345 donors, we found that plasma and serum from most convalescent donors contained IgG antibodies specific to the spike receptor-binding domain (RBD) of SARS-CoV-2, with varying concentrations which correlate with previous disease severity and gender. Anti-RBD IgG plasma concentration significantly correlated with the plasma/serum VN activity against SARS-CoV-2 in vitro., Several hundred millions of people have been diagnosed of coronavirus disease 2019 (COVID-19), causing millions of deaths and a high socioeconomic burden. SARS-CoV-2, the causative agent of COVID-19, induces both specific T- and B-cell responses, being antibodies against the virus detected a few days after infection. Passive immunization with hyperimmune plasma from convalescent patients has been proposed as a potentially useful treatment for COVID-19. Using an in-house quantitative ELISA test, we found that plasma from 177 convalescent donors contained IgG antibodies specific to the spike receptor-binding domain (RBD) of SARS-CoV-2, although at very different concentrations which correlated with previous disease severity and gender. Anti-RBD IgG plasma concentrations significantly correlated with the plasma viral neutralizing activity (VN) against SARS-CoV-2 in vitro. Similar results were found using an independent cohort of serum from 168 convalescent health workers. These results validate an in-house RBD IgG ELISA test in a large cohort of COVID-19 convalescent patients and indicate that plasma from all convalescent donors does not contain a high enough amount of anti-SARS-CoV-2-RBD neutralizing IgG to prevent SARS-CoV-2 infection in vitro. The use of quantitative anti-RBD IgG detection systems might help to predict the efficacy of the passive immunization using plasma from patients recovered from SARS-CoV-2.
- Published
- 2021
15. Inflammatory cell death induced by cytotoxic lymphocytes: a dangerous but necessary liaison
- Author
-
European Commission, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Instituto de Salud Carlos III, Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, de Miguel, Diego [0000-0002-8486-8514], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Uranga Murillo, Iratxe [0000-0001-8411-984X], Hidalgo, Sandra [0000-0003-1629-9978], Santiago, Llipsy [0000-0002-1861-5981], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Arias, Maykel [0000-0002-9730-2210], Pardo, Julián [0000-0003-0154-0730], Miguel, Diego de, Ramírez-Labrada, Ariel, Uranga Murillo, Iratxe, Hidalgo, Sandra, Santiago, Llipsy, Gálvez Buerba, Eva Mª, Arias, Maykel, Pardo, Julián, European Commission, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Instituto de Salud Carlos III, Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, de Miguel, Diego [0000-0002-8486-8514], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Uranga Murillo, Iratxe [0000-0001-8411-984X], Hidalgo, Sandra [0000-0003-1629-9978], Santiago, Llipsy [0000-0002-1861-5981], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Arias, Maykel [0000-0002-9730-2210], Pardo, Julián [0000-0003-0154-0730], Miguel, Diego de, Ramírez-Labrada, Ariel, Uranga Murillo, Iratxe, Hidalgo, Sandra, Santiago, Llipsy, Gálvez Buerba, Eva Mª, Arias, Maykel, and Pardo, Julián
- Abstract
Cytotoxic lymphocytes (CLs), and more specifically Tc and NK cells, are the main executors of cell death in the immune system, playing a key role during both immunosurveillance and immunotherapy. These cells induce regulated cell death (RCD) by different mechanisms, being granular exocytosis and expression of death ligands the most prominent and best characterized ones. Apoptosis, a traditionally considered low-inflammatory type of cell death, has been accepted for years as the paradigm of RCD induced by CLs. However, several recent studies have demonstrated that NK cells and Tc cells can also induce more inflammatory forms of cell death, namely, necroptosis, pyroptosis, and ferroptosis. Activation of these highly inflammatory types of cell death appears to critically contribute to the activation of a successful antitumour immune response. Additionally, the role of specific cell death pathways in immunogenic cell death is still under intense debate, especially considering the interconnections with other inflammatory forms of cell death. These evidences, together with the advent of new cancer immunotherapies, highlight the necessity to deepen our understanding of the link between the cell death triggered by CLs and inflammation. This knowledge will be instrumental to maximize the antitumour potential of immunotherapies, minimizing deleterious effects associated with these treatments. In this review, we will briefly summarize the main features of apoptosis, necroptosis, pyroptosis and ferroptosis, to subsequently discuss the most recent evidences about the role of these RCD pathways during the elimination of cancer cells mediated by CLs and its modulation to increase the efficacy of cancer immunotherapy.
- Published
- 2021
16. Granzyme A inhibition reduces inflammation and increases survival during abdominal sepsis
- Author
-
Garzón, Marcela, Comas, Laura, Santiago, Llipsy, Uranga Murillo, Iratxe, Ramírez-Labrada, Ariel, Gálvez Buerba, Eva Mª, Pardo, Julián, Arias, Maykel, Garzón, Marcela [0000-0001-6778-0636], Comas, Laura [0000-0002-3843-1231], Santiago, Llipsy [0000-0002-1861-5981], Uranga Murillo, Iratxe [0000-0001-8411-984X], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Pardo, Julián [0000-0003-0154-0730], Arias, Maykel [0000-0002-9730-2210], Garzón, Marcela, Comas, Laura, Santiago, Llipsy, Uranga Murillo, Iratxe, Ramírez-Labrada, Ariel, Gálvez Buerba, Eva Mª, Pardo, Julián, and Arias, Maykel
- Subjects
Inflammation ,Sepsis ,Granzyme A ,Cecal ligation and puncture ,Peritonitis - Abstract
3 figuras.-- Póster presentado en el 42º Congreso SEI, Congreso de la Sociedad Española de Inmunología, formato virtual, 24-26 marzo de 2021, Madrid., Peritonitis is one of the most common causes of sepsis. Evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed by NK and T cells, could act as proinflammatory mediator and could play an important role in the pathogenesis of sepsis. Here, we aim to analyze the role and therapeutic potential of GzmA in the pathogenesis of peritoneal sepsis.
- Published
- 2021
17. Ultrastructural analysis and three-dimensional reconstruction of cellular structures involved in SARS-CoV-2 spread
- Author
-
Baselga, Marta, primary, Moreo, Eduardo, additional, Uranga-Murillo, Iratxe, additional, Arias, Maykel, additional, and Junquera, Concepción, additional
- Published
- 2022
- Full Text
- View/download PDF
18. Silver Nanoparticles–Polyethyleneimine-Based Coatings with Antiviral Activity against SARS-CoV-2: A New Method to Functionalize Filtration Media
- Author
-
Baselga, Marta, primary, Uranga-Murillo, Iratxe, additional, de Miguel, Diego, additional, Arias, Maykel, additional, Sebastián, Victor, additional, Pardo, Julián, additional, and Arruebo, Manuel, additional
- Published
- 2022
- Full Text
- View/download PDF
19. Serum active Granzyme A: a new biomarker that contributes to the pathogenesis of peritoneal sepsis
- Author
-
Garzón, Marcela [0000-0001-6778-0636], Sierra Monzón, José L. [0000-0002-8796-2717], Comas, Laura [0000-0002-3843-1231], Uranga Murillo, Iratxe [0000-0001-8411-984X], Morte Romea, Elena [0000-0001-9262-2461], Algarate, Sonia [0000-0002-0036-6630], Ushakova, Tatiana Khaliulina [0000-0002-7930-2129], Luque Gómez, Pilar [0000-0001-7790-409X], Paño, José Ramón [0000-0002-9600-8116], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Arias, Maykel [0000-0002-9730-2210], Pardo, Julián [0000-0003-0154-0730], Garzón, Marcela, Sierra Monzón, José L., Comas, Laura, Uranga Murillo, Iratxe, Morte Romea, Elena, Algarate, Sonia, Ushakova, Tatiana Khaliulina, Seral, Cristina, Luque Gómez, Pilar, Paño, José Ramón, Gálvez Buerba, Eva Mª, Arias, Maykel, Pardo, Julián, Garzón, Marcela [0000-0001-6778-0636], Sierra Monzón, José L. [0000-0002-8796-2717], Comas, Laura [0000-0002-3843-1231], Uranga Murillo, Iratxe [0000-0001-8411-984X], Morte Romea, Elena [0000-0001-9262-2461], Algarate, Sonia [0000-0002-0036-6630], Ushakova, Tatiana Khaliulina [0000-0002-7930-2129], Luque Gómez, Pilar [0000-0001-7790-409X], Paño, José Ramón [0000-0002-9600-8116], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Arias, Maykel [0000-0002-9730-2210], Pardo, Julián [0000-0003-0154-0730], Garzón, Marcela, Sierra Monzón, José L., Comas, Laura, Uranga Murillo, Iratxe, Morte Romea, Elena, Algarate, Sonia, Ushakova, Tatiana Khaliulina, Seral, Cristina, Luque Gómez, Pilar, Paño, José Ramón, Gálvez Buerba, Eva Mª, Arias, Maykel, and Pardo, Julián
- Abstract
[Background] Peritonitis is one of the most common leading cause of sepsis. Recent evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed in NK cells and T cells, could act as a proinflammatory mediator and could play an important role in the pathogenesis of sepsis. This work aims to analyze the role of serum GzmA as a biomarker and therapeutic target in peritoneal sepsis., [Materials/methods] Concentration and enzyme activity of soluble GzmA were sequentially analyzed in serum from healthy donors and patients with peritonitis and were correlated with the Sequential Organ Failure Assessment (SOFA) score. Peritonitis was induced in C57Bl/6 (Wt) and GzmA-KO mice by cecal ligation and puncture (CLP). Mice were treated intraperitoneally with antibiotics and with serpinb6b, a specific GzmA inhibitor, for 5 days. Mouse survival was monitored during 14 days and the levels of serum proinflammatory cytokines and bacterial load in blood and spleen were analyzed at 6 and 24h from CLP., [Results] We have found high levels of GzmA in serum of patient with peritonitis. Most importantly, we observed that GzmA activity in serum correlates with SOFA score, suggesting that active GzmA could play an important role in sepsis development in peritonitis patients and could be a new biomarker of sepsis severity. In order to analyze the therapeutical potential of soluble GzmA in peritoneal sepsis, we used the CLP mouse model. After peritonitis induction, GzmA-KO mice exhibit increased survival compared with Wt mice, which correlated with reduced levels of proinflammatory cytokines in serum. The analysis of bacterial load in blood and spleen showed no differences between Wt and GzmA-KO mice suggesting that GzmA does not play an important role in bacterial control. Treatment with serpinb6b reduced mortality, which correlated with reduced cytokine serum levels in serum, confirming the therapeutical potential of gzmA to treat peritoneal sepsis., [Conclusions] Our findings confirm that soluble GzmA plays an important role in the pathogenesis of sepsis and could be a new therapeutic target and a biomarker for the treatment of peritoneal sepsis.
- Published
- 2020
20. Serum active Granzyme A: a new biomarker that contributes to the pathogenesis of peritoneal sepsis
- Author
-
Garzón, Marcela, Sierra Monzón, José L., Comas, Laura, Uranga Murillo, Iratxe, Morte Romea, Elena, Algarate, Sonia, Ushakova, Tatiana Khaliulina, Seral, Cristina, Luque Gómez, Pilar, Paño, José Ramón, Gálvez Buerba, Eva Mª, Arias, Maykel, Pardo, Julián, Garzón, Marcela, Sierra Monzón, José L., Comas, Laura, Uranga Murillo, Iratxe, Morte Romea, Elena, Algarate, Sonia, Ushakova, Tatiana Khaliulina, Luque Gómez, Pilar, Paño, José Ramón, Gálvez Buerba, Eva Mª, Arias, Maykel, Pardo, Julián, Garzón, Marcela [0000-0001-6778-0636], Sierra Monzón, José L. [0000-0002-8796-2717], Comas, Laura [0000-0002-3843-1231], Uranga Murillo, Iratxe [0000-0001-8411-984X], Morte Romea, Elena [0000-0001-9262-2461], Algarate, Sonia [0000-0002-0036-6630], Ushakova, Tatiana Khaliulina [0000-0002-7930-2129], Luque Gómez, Pilar [0000-0001-7790-409X], Paño, José Ramón [0000-0002-9600-8116], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Arias, Maykel [0000-0002-9730-2210], and Pardo, Julián [0000-0003-0154-0730]
- Subjects
Granzyme ,natural sciences ,Peritoneal sepsis ,Biomarkers - Abstract
of the work presented at the 30th ECCMID 2020, European Congress of Clinical Microbiology and Infectious Diseases, organized by the European Society of Clinical Microbiology and Infectious Diseases (ESCMID).-- www.eccmid.org .-- www.escmid.org.-- Accepted abstract 1508., [Background] Peritonitis is one of the most common leading cause of sepsis. Recent evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed in NK cells and T cells, could act as a proinflammatory mediator and could play an important role in the pathogenesis of sepsis. This work aims to analyze the role of serum GzmA as a biomarker and therapeutic target in peritoneal sepsis., [Materials/methods] Concentration and enzyme activity of soluble GzmA were sequentially analyzed in serum from healthy donors and patients with peritonitis and were correlated with the Sequential Organ Failure Assessment (SOFA) score. Peritonitis was induced in C57Bl/6 (Wt) and GzmA-KO mice by cecal ligation and puncture (CLP). Mice were treated intraperitoneally with antibiotics and with serpinb6b, a specific GzmA inhibitor, for 5 days. Mouse survival was monitored during 14 days and the levels of serum proinflammatory cytokines and bacterial load in blood and spleen were analyzed at 6 and 24h from CLP., [Results] We have found high levels of GzmA in serum of patient with peritonitis. Most importantly, we observed that GzmA activity in serum correlates with SOFA score, suggesting that active GzmA could play an important role in sepsis development in peritonitis patients and could be a new biomarker of sepsis severity. In order to analyze the therapeutical potential of soluble GzmA in peritoneal sepsis, we used the CLP mouse model. After peritonitis induction, GzmA-KO mice exhibit increased survival compared with Wt mice, which correlated with reduced levels of proinflammatory cytokines in serum. The analysis of bacterial load in blood and spleen showed no differences between Wt and GzmA-KO mice suggesting that GzmA does not play an important role in bacterial control. Treatment with serpinb6b reduced mortality, which correlated with reduced cytokine serum levels in serum, confirming the therapeutical potential of gzmA to treat peritoneal sepsis., [Conclusions] Our findings confirm that soluble GzmA plays an important role in the pathogenesis of sepsis and could be a new therapeutic target and a biomarker for the treatment of peritoneal sepsis.
- Published
- 2020
21. Silver nanoparticles–polyethyleneimine-based coatings with antiviral activity against SARS-CoV-2: A new method to functionalize filtration media
- Author
-
Instituto de Salud Carlos III, European Commission, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Miguel, Diego de [0000-0002-8486-8514], Sebastián, Víctor [0000-0002-6873-5244], Arruebo, Manuel [0000-0003-3165-0156], Baselga, Marta, Uranga Murillo, Iratxe, Miguel, Diego de, Arias, Maykel, Sebastián, Víctor, Pardo, Julián, Arruebo, Manuel, Instituto de Salud Carlos III, European Commission, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Miguel, Diego de [0000-0002-8486-8514], Sebastián, Víctor [0000-0002-6873-5244], Arruebo, Manuel [0000-0003-3165-0156], Baselga, Marta, Uranga Murillo, Iratxe, Miguel, Diego de, Arias, Maykel, Sebastián, Víctor, Pardo, Julián, and Arruebo, Manuel
- Abstract
The use of face masks and air purification systems has been key to curbing the transmission of SARS-CoV-2 aerosols in the context of the current COVID-19 pandemic. However, some masks or air conditioning filtration systems are designed to remove large airborne particles or bacteria from the air, being limited their effectiveness against SARS-CoV-2. Continuous research has been aimed at improving the performance of filter materials through nanotechnology. This article presents a new low-cost method based on electrostatic forces and coordination complex formation to generate antiviral coatings on filter materials using silver nanoparticles and polyethyleneimine. Initially, the AgNPs synthesis procedure was optimized until reaching a particle size of 6.2 ± 2.6 nm, promoting a fast ionic silver release due to its reduced size, obtaining a stable colloid over time and having reduced size polydispersity. The stability of the binding of the AgNPs to the fibers was corroborated using polypropylene, polyester-viscose, and polypropylene-glass spunbond mats as substrates, obtaining very low amounts of detached AgNPs in all cases. Under simulated operational conditions, a material loss less than 1% of nanostructured silver was measured. SEM micrographs demonstrated high silver distribution homogeneity on the polymer fibers. The antiviral coatings were tested against SARS-CoV-2, obtaining inactivation yields greater than 99.9%. We believe our results will be beneficial in the fight against the current COVID-19 pandemic and in controlling other infectious airborne pathogens
- Published
- 2022
22. Adoptive NK Cell Transfer as a Treatment in Colorectal Cancer Patients: Analyses of Tumour Cell Determinants Correlating With Efficacy In Vitro and In Vivo
- Author
-
Lanuza, Pilar M., primary, Alonso, M. Henar, additional, Hidalgo, Sandra, additional, Uranga-Murillo, Iratxe, additional, García-Mulero, Sandra, additional, Arnau, Raquel, additional, Santos, Cristina, additional, Sanjuan, Xavier, additional, Santiago, Llipsy, additional, Comas, Laura, additional, Redrado, Sergio, additional, Pazo-Cid, Roberto, additional, Agustin-Ferrández, M. Jose, additional, Jaime-Sánchez, Paula, additional, Pesini, Cecilia, additional, Gálvez, Eva M., additional, Ramírez-Labrada, Ariel, additional, Arias, Maykel, additional, Sanz-Pamplona, Rebeca, additional, and Pardo, Julián, additional
- Published
- 2022
- Full Text
- View/download PDF
23. All About (NK Cell-Mediated) Death in Two Acts and an Unexpected Encore: Initiation, Execution and Activation of Adaptive Immunity
- Author
-
Ramírez-Labrada, Ariel, primary, Pesini, Cecilia, additional, Santiago, Llipsy, additional, Hidalgo, Sandra, additional, Calvo-Pérez, Adanays, additional, Oñate, Carmen, additional, Andrés-Tovar, Alejandro, additional, Garzón-Tituaña, Marcela, additional, Uranga-Murillo, Iratxe, additional, Arias, Maykel A., additional, Galvez, Eva M., additional, and Pardo, Julián, additional
- Published
- 2022
- Full Text
- View/download PDF
24. Inflammatory cell death induced by cytotoxic lymphocytes: a dangerous but necessary liaison
- Author
-
Miguel, Diego de, Ramírez-Labrada, Ariel, Uranga Murillo, Iratxe, Hidalgo, Sandra, Santiago, Llipsy, Gálvez Buerba, Eva Mª, Arias, Maykel, Pardo, Julián, European Commission, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Instituto de Salud Carlos III, Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, de Miguel, Diego [0000-0002-8486-8514], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Uranga Murillo, Iratxe [0000-0001-8411-984X], Hidalgo, Sandra [0000-0003-1629-9978], Santiago, Llipsy [0000-0002-1861-5981], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Arias, Maykel [0000-0002-9730-2210], and Pardo, Julián [0000-0003-0154-0730]
- Subjects
Inflammation ,Immunosurveillance ,Necroptosis ,Pyroptosis ,Ferroptosis ,Apoptosis ,Immunotherapy ,Immunogenic cell death - Abstract
2 figures. Cytotoxic lymphocytes (CLs), and more specifically Tc and NK cells, are the main executors of cell death in the immune system, playing a key role during both immunosurveillance and immunotherapy. These cells induce regulated cell death (RCD) by different mechanisms, being granular exocytosis and expression of death ligands the most prominent and best characterized ones. Apoptosis, a traditionally considered low-inflammatory type of cell death, has been accepted for years as the paradigm of RCD induced by CLs. However, several recent studies have demonstrated that NK cells and Tc cells can also induce more inflammatory forms of cell death, namely, necroptosis, pyroptosis, and ferroptosis. Activation of these highly inflammatory types of cell death appears to critically contribute to the activation of a successful antitumour immune response. Additionally, the role of specific cell death pathways in immunogenic cell death is still under intense debate, especially considering the interconnections with other inflammatory forms of cell death. These evidences, together with the advent of new cancer immunotherapies, highlight the necessity to deepen our understanding of the link between the cell death triggered by CLs and inflammation. This knowledge will be instrumental to maximize the antitumour potential of immunotherapies, minimizing deleterious effects associated with these treatments. In this review, we will briefly summarize the main features of apoptosis, necroptosis, pyroptosis and ferroptosis, to subsequently discuss the most recent evidences about the role of these RCD pathways during the elimination of cancer cells mediated by CLs and its modulation to increase the efficacy of cancer immunotherapy. Work in the JP laboratory is funded by the FEDER (Fondo Europeo de Desarrollo Regional, Gobierno de Aragón, Group B29_17R), Ministerio de Ciencia, Innovación e Universidades (MCNU), Health National Institute Carlos III, Agencia Estatal de Investigación (SAF2017-83120-C2-1-R; PID2020-113963RB-I00), Fundación Inocente Inocente, ASPANOA and Carrera de la Mujer de Monzón. EMG is funded by Agencia Estatal de Investigación (SAF2017-83120-C2-1-R and PID2020-113963RB-I00). DDM is supported by a postdoctoral fellowship ‘Sara Borrell’, LS by a PhD fellowship (FPI) from the Ministry of Science, Innovation and Universities. IUM and SH are supported by a PhD fellowship from Aragon Government. MA is supported by a postdoctoral fellowship ‘Juan de la Cierva-formación’ from the Ministry of Science, Innovation and Universities, and JP is supported by the ARAID Foundation.
- Published
- 2021
25. Integrated analysis of circulating immune cellular and soluble mediators reveals specific COVID19 signatures at hospital admission with utility for prediction of clinical outcomes
- Author
-
Uranga-Murillo, Iratxe, primary, Morte, Elena, additional, Hidalgo, Sandra, additional, Pesini, Cecilia, additional, García-Mulero, Sandra, additional, Sierra, Jose L., additional, Santiago, Llipsy, additional, Arias, Maykel, additional, De Miguel, Diego, additional, Encabo-Berzosa, María del Mar, additional, Gracia-Tello, Borja, additional, Sanz-Pamplona, Rebeca, additional, Martinez-Lostao, Luis, additional, Galvez, Eva M., additional, Paño-Pardo, Jose R., additional, Ramirez-Labrada, Ariel, additional, and Pardo, Julian, additional
- Published
- 2022
- Full Text
- View/download PDF
26. Epitope spreading driven by the joint action of CART cells and pharmacological STING stimulation counteracts tumor escape via antigen-loss variants
- Author
-
Conde, Enrique, primary, Vercher, Enric, additional, Soria-Castellano, Marta, additional, Suarez-Olmos, Jesús, additional, Mancheño, Uxua, additional, Elizalde, Edurne, additional, Rodriguez, M Luis, additional, Glez-Vaz, Javier, additional, Casares, Noelia, additional, Rodríguez-García, Estefanía, additional, Hommel, Mirja, additional, González-Aseguinolaza, Gloria, additional, Uranga-Murillo, Iratxe, additional, Pardo, Julian, additional, Alkorta, Gorka, additional, Melero, Ignacio, additional, Lasarte, Juan, additional, and Hervas-Stubbs, Sandra, additional
- Published
- 2021
- Full Text
- View/download PDF
27. Concentration of IgG against the Spike Receptor-Binding Domain predicts the viral neutralization activity of convalescent plasma and serum against SARS-CoV-2
- Author
-
Santiago, Llipsy, Uranga Murillo, Iratxe, Arias, Maykel, Moreo, Eduardo, Hidalgo, Sandra, Gálvez Buerba, Eva Mª, Ramírez-Labrada, Ariel, Miguel, Diego de, Paño, José Ramón, and Pardo, Julián
- Subjects
Coronavirus ,Convalescent plasma ,SARS-CoV-2 ,IgG ,ELISA ,Antibodies - Abstract
Poster presentado en el 42º SEI Congreso de la Sociedad Española de Inmunología, 24-26 marzo 2021, Madrid, formato virtual., SARS-CoV-2 is the virus responsible for the Covid-19 pandemic, due to its rapid propagation and the initial lack of vaccines or appropriate treatments. Nowadays, despite different vaccines available, the main treatments are palliative, focused on supplemental oxygen and anti-inflammatory therapy. Passive immunization could be an effective and economic treatment once standarized. It consists of the transfer of pathogen-specific antibodies to patients whose immune system has not originated a response yet. The donors’ antibodies neutralize and attenuate pathogen replication. Besides, an antibody against the Receptor-Binding Domain of Spike (RBD) would block the interaction of the virus with ACE2 and its entry in the cell. Here, an in-house RBG IgG ELISA test has been validated using a cohort of more than 320 samples of convalescent plasma and serum and adapted to quantify the concentration of plasma RBD IgG and its correlation with the SARS-CoV-2 neutralizing activity in vitro.
- Published
- 2021
28. TIM3, LAG3, CXCL10 and GzmA reveal as main hallmarks of inflammatory profiles in Covid-19 patiens
- Author
-
Hidalgo, Sandra, Morte Romea, Elena, Pesini, Cecilia, Uranga Murillo, Iratxe, Sierra Monzón, José L., Martínez Lázaro, Beatriz, Paño, José Ramón, Gálvez Buerba, Eva Mª, and Pardo, Julián
- Subjects
Inflammation ,Immune ,SARS-CoV-2 ,CXCL10 ,Granzyme A ,COVID-19 ,TIM3 ,Cytokine ,LAG3 - Abstract
Comunicación oral presentada en el 42º SEI Congreso de la Sociedad Española de Inmunología, 24-26 marzo 2021, Madrid, formato virtual.
- Published
- 2021
29. Determination of the concentration of igg against the spike receptor-binding domain that predicts the viral neutralizing activity of convalescent plasma and serum against sars-cov-2
- Author
-
Santiago, Llipsy, Uranga-Murillo, Iratxe, Arias, Maykel, González-Ramírez, Andrés Manuel, Macías-León, Javier, Moreo, Eduardo, Redrado, Sergio, García-García, Ana, Taleb, Víctor, Lira-Navarrete, Erandi, Hurtado-Guerrero, Ramón, Aguilo, Nacho, Encabo-Berzosa, Maria Del Mar, Hidalgo, Sandra, Galvez, Eva M., Ramirez-Labrada, Ariel, de Miguel, Diego, Benito, Rafael, Miranda, Patricia, Fernández, Antonio, Domingo, José María, Serrano, Laura, Yuste, Cristina, Villanueva-Saz, Sergio, Paño-Pardo, José Ramón, Pardo, Julián, Santiago, Llipsy, Uranga-Murillo, Iratxe, Arias, Maykel, González-Ramírez, Andrés Manuel, Macías-León, Javier, Moreo, Eduardo, Redrado, Sergio, García-García, Ana, Taleb, Víctor, Lira-Navarrete, Erandi, Hurtado-Guerrero, Ramón, Aguilo, Nacho, Encabo-Berzosa, Maria Del Mar, Hidalgo, Sandra, Galvez, Eva M., Ramirez-Labrada, Ariel, de Miguel, Diego, Benito, Rafael, Miranda, Patricia, Fernández, Antonio, Domingo, José María, Serrano, Laura, Yuste, Cristina, Villanueva-Saz, Sergio, Paño-Pardo, José Ramón, and Pardo, Julián
- Abstract
Several hundred millions of people have been diagnosed of coronavirus disease 2019 (COVID-19), causing millions of deaths and a high socioeconomic burden. SARS-CoV-2, the causative agent of COVID-19, induces both specific T-and B-cell responses, being antibodies against the virus detected a few days after infection. Passive immunization with hyperimmune plasma from convalescent patients has been proposed as a potentially useful treatment for COVID-19. Using an in-house quantitative ELISA test, we found that plasma from 177 convalescent donors contained IgG antibodies specific to the spike receptor-binding domain (RBD) of SARS-CoV-2, although at very different concentrations which correlated with previous disease severity and gender. Anti-RBD IgG plasma concentrations significantly correlated with the plasma viral neutralizing activity (VN) against SARS-CoV-2 in vitro. Similar results were found using an independent cohort of serum from 168 convalescent health workers. These results validate an in-house RBD IgG ELISA test in a large cohort of COVID-19 convalescent patients and indicate that plasma from all convalescent donors does not contain a high enough amount of anti-SARS-CoV-2-RBD neutralizing IgG to prevent SARS-CoV-2 infection in vitro. The use of quantitative anti-RBD IgG detection systems might help to predict the efficacy of the passive immunization using plasma from patients recovered from SARS-CoV-2.
- Published
- 2021
30. Analysis of the inflammatory cytokine and cell immunity profile in blood that predicts disease severity in COVID-19 patients
- Author
-
Hidalgo, Sandra, Morte Romea, Elena, Pesini, Cecilia, Uranga Murillo, Iratxe, Sierra Monzón, José L., Martínez Lázaro, Beatriz, Paño, José Ramón, Gálvez Buerba, Eva Mª, Pardo, Julián, Hidalgo, Sandra, Morte Romea, Elena, Pesini, Cecilia, Uranga Murillo, Iratxe, Sierra Monzón, José L., Martínez Lázaro, Beatriz, Paño, José Ramón, Gálvez Buerba, Eva Mª, and Pardo, Julián
- Published
- 2021
31. Efecto de nanopartículas de oro sobre el microambiente inmunológico tumoral
- Author
-
Comas, Laura, Uranga Murillo, Iratxe, Artiga, Álvaro, Serrano, Inés, Arias, Maykel, Fuente, Jesús M. de la, Pardo, Julián, and Gálvez Buerba, Eva Mª
- Subjects
Cancer microenvironment ,Inflammation ,Inflamación ,Microambiente tumoral ,Gold nanoparticles ,Nanopartículas de oro ,Nanopartículas poliméricas ,Polymeric nanoparticles - Abstract
Trabajo presentado en el III Workshop de NanoOncología, 14-15 de Enero de 2020, en la Facultad de Medicina de la Universidad de Zaragoza (España).
- Published
- 2020
32. Nuevas dianas terapéuticas en sepsis
- Author
-
Arias, Maykel, Garzón, Marcela, Comas, Laura, Uranga Murillo, Iratxe, Santiago, Llipsy, Pardo, Julián, and Gálvez Buerba, Eva Mª
- Subjects
Granzimas ,Biomarcadores ,Sepsis ,Peritoneal sepsis - Abstract
Trabajo presentado en el III Workshop NanoOncología en Zaragoza, celebrado del 14 al 15 de enero de 2020, en la Facultad de Medicina de la Universidad de Zaragoza.
- Published
- 2020
33. Determination of the Concentration of IgG against the Spike Receptor-Binding Domain That Predicts the Viral Neutralizing Activity of Convalescent Plasma and Serum against SARS-CoV-2
- Author
-
Santiago, Llipsy, primary, Uranga-Murillo, Iratxe, additional, Arias, Maykel, additional, González-Ramírez, Andrés Manuel, additional, Macías-León, Javier, additional, Moreo, Eduardo, additional, Redrado, Sergio, additional, García-García, Ana, additional, Taleb, Víctor, additional, Lira-Navarrete, Erandi, additional, Hurtado-Guerrero, Ramón, additional, Aguilo, Nacho, additional, del Mar Encabo-Berzosa, Maria, additional, Hidalgo, Sandra, additional, Galvez, Eva M., additional, Ramirez-Labrada, Ariel, additional, de Miguel, Diego, additional, Benito, Rafael, additional, Miranda, Patricia, additional, Fernández, Antonio, additional, Domingo, José María, additional, Serrano, Laura, additional, Yuste, Cristina, additional, Villanueva-Saz, Sergio, additional, Paño-Pardo, José Ramón, additional, and Pardo, Julián, additional
- Published
- 2021
- Full Text
- View/download PDF
34. Granzyme A inhibition reduces inflammation and increases survival during abdominal sepsis
- Author
-
Garzón-Tituaña, Marcela, primary, Sierra-Monzón, José L, additional, Comas, Laura, additional, Santiago, Llipsy, additional, Khaliulina-Ushakova, Tatiana, additional, Uranga-Murillo, Iratxe, additional, Ramirez-Labrada, Ariel, additional, Tapia, Elena, additional, Morte-Romea, Elena, additional, Algarate, Sonia, additional, Couty, Ludovic, additional, Camerer, Eric, additional, Bird, Phillip I, additional, Seral, Cristina, additional, Luque, Pilar, additional, Paño-Pardo, José R, additional, Galvez, Eva M, additional, Pardo, Julián, additional, and Arias, Maykel, additional
- Published
- 2021
- Full Text
- View/download PDF
35. Biological relevance of Granzymes A and K during E. coli sepsis
- Author
-
Uranga-Murillo, Iratxe, primary, Tapia, Elena, additional, Garzón-Tituaña, Marcela, additional, Ramirez-Labrada, Ariel, additional, Santiago, Llipsy, additional, Pesini, Cecilia, additional, Esteban, Patricia, additional, Roig, Francisco J, additional, Galvez, Eva M, additional, Bird, Phillip I, additional, Pardo, Julián, additional, and Arias, Maykel, additional
- Published
- 2021
- Full Text
- View/download PDF
36. First-described recently discovered non-toxic vegetal-derived furocoumarin preclinical efficacy against SARS-CoV-2: a promising antiviral herbal drug
- Author
-
Galindo-Cardiel, Iván José, primary, Núñez, Adriana Toledo, additional, Fernández, María Celaya, additional, Ramírez Labrada, Ariel, additional, Uranga-Murillo, Iratxe, additional, Cabrero, Maykel Arias, additional, Pardo, Julian, additional, and Panzeri, Ezio, additional
- Published
- 2020
- Full Text
- View/download PDF
37. Extracellular Granzyme A Promotes Colorectal Cancer Development by Enhancing Gut Inflammation
- Author
-
Santiago, Llipsy, primary, Castro, Marta, additional, Sanz-Pamplona, Rebeca, additional, Garzón, Marcela, additional, Ramirez-Labrada, Ariel, additional, Tapia, Elena, additional, Moreno, Víctor, additional, Layunta, Elena, additional, Gil-Gómez, Gabriel, additional, Garrido, Marta, additional, Peña, Raúl, additional, Lanuza, Pilar M., additional, Comas, Laura, additional, Jaime-Sanchez, Paula, additional, Uranga-Murillo, Iratxe, additional, del Campo, Rosa, additional, Pelegrín, Pablo, additional, Camerer, Eric, additional, Martínez-Lostao, Luis, additional, Muñoz, Guillermo, additional, Uranga, José A., additional, Alcalde, Anabel, additional, Galvez, Eva M., additional, Ferrandez, Angel, additional, Bird, Phillip I., additional, Metkar, Sunil, additional, Arias, Maykel A., additional, and Pardo, Julian, additional
- Published
- 2020
- Full Text
- View/download PDF
38. Granzyme A inhibition reduces inflammation and increases survival during abdominal sepsis
- Author
-
Garzón-Tituaña, Marcela, primary, Sierra-Monzón, José L, additional, Comas, Laura, additional, Santiago, Llipsy, additional, Khaliulina-Ushakova, Tatiana, additional, Uranga-Murillo, Iratxe, additional, Ramirez-Labrada, Ariel, additional, Tapia, Elena, additional, Morte-Romea, Elena, additional, Algarate, Sonia, additional, Couty, Ludovic, additional, Camerer, Eric, additional, Bird, Phillip I, additional, Seral, Cristina, additional, Luque, Pilar, additional, Paño-Pardo, José R, additional, Galvez, Eva M, additional, Pardo, Julián, additional, and Arias, Maykel, additional
- Published
- 2020
- Full Text
- View/download PDF
39. Cell death induced by cytotoxic CD8+T cells is immunogenic and primes caspase-3–dependent spread immunity against endogenous tumor antigens
- Author
-
Jaime-Sanchez, Paula, primary, Uranga-Murillo, Iratxe, additional, Aguilo, Nacho, additional, Khouili, Sofia C, additional, Arias, Maykel A, additional, Sancho, David, additional, and Pardo, Julian, additional
- Published
- 2020
- Full Text
- View/download PDF
40. Serpinb6b inhibits Granzyme A-mediated macrophage inflammatory response and improves sepsis and inflammatory colorectal cancer
- Author
-
Santiago, Llipsy, Garzón, Marcela, Comas, Laura, Uranga Murillo, Iratxe, Ramírez-Labrada, Ariel, Arias, Maykel, Bird, Phillip I., Gálvez Buerba, Eva Mª, and Pardo, Julián
- Subjects
Inflammation ,Sepsis ,Granzyme A ,Serpinb6b ,Peritonitis ,Colorectal cancer - Abstract
Oral presentation made at the Serpins2019 Conference, held in Sevilla (Spain), 19th – 22nd September 2019.
- Published
- 2019
41. Modulation of inflammation in the tumour microenvironment by gold and polymeric nanoparticles
- Author
-
Comas, Laura, Uranga Murillo, Iratxe, Artiga, Álvaro, Serrano, Inés, Arias, Maykel, Martínez de la Fuente, Jesús, Pardo, Julián, and Gálvez Buerba, Eva Mª
- Subjects
Cancer microenvironment ,Inflammation ,Gold nanoparticles ,Polymeric nanoparticles - Abstract
of the poster presented at the III SYMPOSIUM IMMUNOTHERAPY CANCER, held in Zaragoza, November 7th, 2019., [Introduction]: The number of patients benefiting from Immune Checkpoint Inhibitors (ICIs) is still relatively low, restricted to specific tumour types; those presenting specific inflammatory immune cell populations at tumour microenvironment: the so-called hot tumours. Development of protocols to selectively modulate inflammation and increase tumour sensitivity to ICIs is required. Some nanoparticles (NPs) are able to modulate the immune response. Yet, their effect on tumour microenvironment and inflammation has not been explored. [Aim]: test the effect of NPs of different composition on the inflammatory response in myeloid cells in vitro and in tumours in vivo., [Methods]: M1/M2 macrophages and dendritic cells (DCs) were differentiated from mouse bone marrow. Gold and chitosan NPs (GNPs or CNCs) were synthetized and characterised. Production of inflammatory cytokines in myeloid cells and the mechanism involved were analysed in vitro. Immunomodulation in vivo was analysed in the B16 melanoma model., [Results]: GNPs are not cytotoxic in vitro at any concentration tested, in contrast to CNCs, that present cytotoxicity at high concentration. Both GNPs and CNCs were able to induce the release of TNFα and IL6 in macrophages in vitro, and, in combination with LPS, they also induced IL1β. The latter depended on the canonical caspase-1-inflammasome. NPs also induced DC maturation in vitro. GNPs increased in vivo LPS immunomodulation in tumours, modifying the tumour growth and the microenvironment, including cell populations and the profile of inflammatory cytokines., [Conclusions]: GNPs and CNCs are able to modulate the inflammatory response in tumour microenvironment in vitro and in vivo. Further studies will required to analyse if they modulate the sensitivity of tumour to ICIs.
- Published
- 2019
42. Antigen-specific primed cytotoxic T cells eliminate tumour cells in vivo and prevent tumour development, regardless of the presence of anti-apoptotic mutations conferring drug resistance
- Author
-
Ministerio de Economía y Competitividad (España), European Commission, Universidad de Zaragoza, Gobierno de Aragón, Ministerio de Educación, Cultura y Deporte (España), Fundación Banco Santander, ARAID Foundation, Ministerio de Ciencia e Innovación (España), Agencia Estatal de Investigación (España), Ministerio de Ciencia, Innovación y Universidades (España), Jaime-Sánchez, Paula, Catalán, Elena, Uranga Murillo, Iratxe, Aguiló, Nacho, Santiago, Llipsy, Lanuza, Pilar M., Miguel, Diego de, Arias, Maykel, Pardo, Julián, Ministerio de Economía y Competitividad (España), European Commission, Universidad de Zaragoza, Gobierno de Aragón, Ministerio de Educación, Cultura y Deporte (España), Fundación Banco Santander, ARAID Foundation, Ministerio de Ciencia e Innovación (España), Agencia Estatal de Investigación (España), Ministerio de Ciencia, Innovación y Universidades (España), Jaime-Sánchez, Paula, Catalán, Elena, Uranga Murillo, Iratxe, Aguiló, Nacho, Santiago, Llipsy, Lanuza, Pilar M., Miguel, Diego de, Arias, Maykel, and Pardo, Julián
- Abstract
Cytotoxic CD8+ T (Tc) cells are the main executors of transformed and cancer cells during cancer immunotherapy. The latest clinical results evidence a high efficacy of novel immunotherapy agents that modulate Tc cell activity against bad prognosis cancers. However, it has not been determined yet whether the efficacy of these treatments can be affected by selection of tumoural cells with mutations in the cell death machinery, known to promote drug resistance and cancer recurrence. Here, using a model of prophylactic tumour vaccination based on the LCMV-gp33 antigen and the mouse EL4 T lymphoma, we analysed the molecular mechanism employed by Tc cells to eliminate cancer cells in vivo and the impact of mutations in the apoptotic machinery on tumour development. First of all, we found that Tc cells, and perf and gzmB are required to efficiently eliminate EL4.gp33 cells after LCMV immunisation during short-term assays (1-4 h), and to prevent tumour development in the long term. Furthermore, we show that antigen-pulsed chemoresistant EL4 cells overexpressing Bcl-XL or a dominant negative form of caspase-3 are specifically eliminated from the peritoneum of infected animals, as fast as parental EL4 cells. Notably, antigen-specific Tc cells control the tumour growth of the mutated cells, as efficiently as in the case of parental cells. Altogether, expression of the anti-apoptotic mutations does not confer any advantage for tumour cells neither in the short-term survival nor in long-term tumour formation. Although the mechanism involved in the elimination of the apoptosis-resistant tumour cells is not completely elucidated, neither necroptosis nor pyroptosis seem to be involved. Our results provide the first experimental proof that chemoresistant cancer cells with mutations in the main cell death pathways are efficiently eliminated by Ag-specific Tc cells in vivo during immunotherapy and, thus, provide the molecular basis to treat chemoresistant cancer cells with CD8 Tc-base
- Published
- 2018
43. Cleavage of cFLIP restrains cell death during viral infection and tissue injury and favors tissue repair.
- Author
-
Lagunas, Kristel Martinez, Savcigil, Deniz Pinar, Zrilic, Matea, Fraile, Carlos Carvajal, Craxton, Andrew, Self, Emily, Uranga-Murillo, Iratxe, de Miguel, Diego, Arias, Maykel, Willenborg, Sebastian, Piekarek, Michael, Albert, Marie Christine, Nugraha, Kalvin, Lisewski, Ina, Janakova, Erika, Igual, Natalia, Tonnus, Wulf, Hildebrandt, Ximena, Ibrahim, Mohammed, and Ballegeer, Marlies
- Subjects
- *
CELL death , *VIRUS diseases , *SKIN regeneration , *DEATH receptors , *SOFT tissue injuries , *INTERFERON gamma , *BONE marrow cells - Abstract
The article focuses on a study conducted to analyze the physiological role of cellular FLICE-like inhibitory protein (cFLIP) cleavage by Caspase-8 in preventing cell death in a variety of pathological circumstances. It found that a cleavage-resistant cFLIP mutant, CflipD377A, showed greater vulnerability to the SARS-CoV, slowed skin wound healing, and increased Sharpin deficiency-induced tissue damage.
- Published
- 2023
- Full Text
- View/download PDF
44. All about (nk cell-mediated) death in two acts and an unexpected encore: initiation, execution and activation of adaptive immunity
- Author
-
Ariel Ramírez-Labrada, Cecilia Pesini, Llipsy Santiago, Sandra Hidalgo, Adanays Calvo-Pérez, Carmen Oñate, Alejandro Andrés-Tovar, Marcela Garzón-Tituaña, Iratxe Uranga-Murillo, Maykel A. Arias, Eva M. Galvez, Julián Pardo, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Gobierno de Aragón, Agencia Estatal de Investigación (España), Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, ARAID Foundation, Asociación Española Contra el Cáncer, Ministerio de Ciencia, Innovación y Universidades (España), Ramírez-Labrada, Ariel, Pesini, Cecilia, Santiago, Llipsy, Hidalgo, Sandra, Uranga Murillo, Iratxe, Arias, Maykel, Gálvez Buerba, Eva Mª, Pardo, Julián, Ramírez-Labrada, Ariel [0000-0002-3888-7036], Pesini, Cecilia [0000-0002-8707-2722], Santiago, Llipsy [0000-0002-1861-5981], Hidalgo, Sandra [0000-0003-1629-9978], Uranga Murillo, Iratxe [0000-0001-8411-984X], Arias, Maykel [0000-0002-9730-2210], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], and Pardo, Julián [0000-0003-0154-0730]
- Subjects
Cytotoxicity, Immunologic ,Cell death ,Perforin ,Death receptors ,Immunology ,NK cells ,Adaptive Immunity ,Ligands ,Granzymes ,Killer Cells, Natural ,Neoplasms ,Tumor Microenvironment ,Humans ,Immunology and Allergy ,Immunological cell death ,Granule exocytosis - Abstract
3 figures., NK cells are key mediators of immune cell-mediated cytotoxicity toward infected and transformed cells, being one of the main executors of cell death in the immune system. NK cells recognize target cells through an array of inhibitory and activating receptors for endogenous or exogenous pathogen-derived ligands, which together with adhesion molecules form a structure known as immunological synapse that regulates NK cell effector functions. The main and best characterized mechanisms involved in NK cell-mediated cytotoxicity are the granule exocytosis pathway (perforin/granzymes) and the expression of death ligands. These pathways are recognized as activators of different cell death programmes on the target cells leading to their destruction. However, most studies analyzing these pathways have used pure recombinant or native proteins instead of intact NK cells and, thus, extrapolation of the results to NK cell-mediated cell death might be difficult. Specially, since the activation of granule exocytosis and/or death ligands during NK cell-mediated elimination of target cells might be influenced by the stimulus received from target cells and other microenvironment components, which might affect the cell death pathways activated on target cells. Here we will review and discuss the available experimental evidence on how NK cells kill target cells, with a special focus on the different cell death modalities that have been found to be activated during NK cell-mediated cytotoxicity; including apoptosis and more inflammatory pathways like necroptosis and pyroptosis. In light of this new evidence, we will develop the new concept of cell death induced by NK cells as a new regulatory mechanism linking innate immune response with the activation of tumour adaptive T cell responses, which might be the initiating stimulus that trigger the cancer-immunity cycle. The use of the different cell death pathways and the modulation of the tumour cell molecular machinery regulating them might affect not only tumour cell elimination by NK cells but, in addition, the generation of T cell responses against the tumour that would contribute to efficient tumour elimination and generate cancer immune memory preventing potential recurrences., This research was supported by CIBER -Consorcio Centro de Investigación Biomédica en Red- (CB 2021), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación and Unión Europea – NextGenerationEU, FEDER (Fondo Europeo de Desarrollo Regional, Gobierno de Aragón (Group B29_20R), Ministerio de Ciencia, Innovación e Universidades (MCNU), Agencia Estatal de Investigación (SAF2017-83120-C2-1-R; PID2020-113963RB-I00), ASPANOA, Donación Javier Saura Carceller, and Carrera de la Mujer de Monzón. JP was supported by Fundacion Aragon I+D (ARAID). MA was granted by a Juan de la Cierva contract of Spanish Ministry of Economy and Competitiveness (IJC2019-039192-I). LS was granted by a Juan de la Cierva contract of Spanish Ministry of Economy and Competitiveness (FJC2020-046181-I). IU-M and SH are supported by a PhD fellowship from Aragon Government, CP by a PhD fellowship from Fundación Científica de la Asociación Española Contra el Cáncer (FC AECC), MG-T by a PhD fellowship (FPI) from the Ministry of Science, Innovation and Universities. CO was granted by AEI -Agencia Estatal de Investigación- (PTA2020-018510-I / AEI / 10.13039/501100011033).
- Published
- 2022
45. Adoptive NK Cell Transfer as a Treatment in Colorectal Cancer Patients: Analyses of Tumour Cell Determinants Correlating With Efficacy In Vitro and In Vivo
- Author
-
Pilar M. Lanuza, M. Henar Alonso, Sandra Hidalgo, Iratxe Uranga-Murillo, Sandra García-Mulero, Raquel Arnau, Cristina Santos, Xavier Sanjuan, Llipsy Santiago, Laura Comas, Sergio Redrado, Roberto Pazo-Cid, M. Jose Agustin-Ferrández, Paula Jaime-Sánchez, Cecilia Pesini, Eva M. Gálvez, Ariel Ramírez-Labrada, Maykel Arias, Rebeca Sanz-Pamplona, Julián Pardo, Aspanoa, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Fundación Inocente Inocente, Asociación Carrera de la Mujer Ciudad de Monzón, Gobierno de Aragón, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), ARAID Foundation, Institut Català d'Oncologia, Ministerio de Economía, Industria y Competitividad (España), Centro de Investigación Biomédica en Red Epidemiología y Salud Pública (España), Agència de Gestió d'Ajuts Universitaris i de Recerca, European Cooperation in Science and Technology, Lanuza, Pilar M., Alonso, M. Henar, Hidalgo, Sandra, Uranga Murillo, Iratxe, García-Mulero, Sandra, Sanjuan, Xavier, Santiago, Llipsy, Comas, Laura, Redrado, Sergio, Pazo-Cid, R., Jaime Sánchez, Paula, Pesini, Cecilia, Gálvez Buerba, Eva Mª, Ramírez-Labrada, Ariel, Arias, Maykel, Sanz-Pamplona, Rebeca, Pardo, Julián, Lanuza, Pilar M. [0000-0001-7328-2094], Alonso, M. Henar [0000-0003-0285-5451], Hidalgo, Sandra [0000-0003-1629-9978], Uranga Murillo, Iratxe [0000-0001-8411-984X], García-Mulero, Sandra [0000-0003-4931-1267], Sanjuan, Xavier [0000-0002-5253-822X], Santiago, Llipsy [0000-0002-1861-5981], Comas, Laura [0000-0002-3843-1231], Redrado, Sergio [0000-0002-8404-0012], Pazo-Cid, R. [0000-0002-8026-7391], Jaime Sánchez, Paula [0000-0002-8731-4269], Pesini, Cecilia [0000-0002-8707-2722], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Arias, Maykel [0000-0002-9730-2210], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], and Pardo, Julián [0000-0003-0154-0730]
- Subjects
Cancer biomarker ,Càncer colorectal ,Immunology ,Immune checkpoints ,Immunoteràpia ,HLA-I ,Immunology and Allergy ,Immunotheraphy ,NK cell immunotherapy ,Colorectal cancer ,Tumour immune microenvironment - Abstract
6 figures.-- Supplementary material available., Background: Colorectal cancer (CRC) is a heterogeneous disease with variable mutational profile and tumour microenvironment composition that influence tumour progression and response to treatment. While chemoresistant and poorly immunogenic CRC remains a challenge, the development of new strategies guided by biomarkers could help stratify and treat patients. Allogeneic NK cell transfer emerges as an alternative against chemoresistant and poorly immunogenic CRC., Methods: NK cell-related immunological markers were analysed by transcriptomics and immunohistochemistry in human CRC samples and correlated with tumour progression and overall survival. The anti-tumour ability of expanded allogeneic NK cells using a protocol combining cytokines and feeder cells was analysed in vitro and in vivo and correlated with CRC mutational status and the expression of ligands for immune checkpoint (IC) receptors regulating NK cell activity., Results: HLA-I downmodulation and NK cell infiltration correlated with better overall survival in patients with a low-stage (II) microsatellite instability-high (MSI-H) CRC, suggesting a role of HLA-I as a prognosis biomarker and a potential benefit of NK cell immunotherapy. Activated allogeneic NK cells were able to eliminate CRC cultures without PD-1 and TIM-3 restriction but were affected by HLA-I expression. In vivo experiments confirmed the efficacy of the therapy against both HLA+ and HLA− CRC cell lines. Concomitant administration of pembrolizumab failed to improve tumour control., Conclusions: Our results reveal an immunological profile of CRC tumours in which immunogenicity (MSI-H) and immune evasion mechanisms (HLA downmodulation) favour NK cell immunosurveillance at early disease stages. Accordingly, we have shown that allogeneic NK cell therapy can target tumours expressing mutations conferring poor prognosis regardless of the expression of T cell-related inhibitory IC ligands. Overall, this study provides a rationale for a new potential basis for CRC stratification and NK cell-based therapy., Work in the JP laboratory is funded by ASPANOA, CIBER (CB 2021; Instituto de Salud Carlos III, Ministerio de Ciencia, Innovación and Union Europea.NextGenerationEU), Fundacion Inocente, Carrera de la Mujer Monzón, FEDER/Gobierno de Aragón (Group B29_17R), and Ministerio de Ciencia, Innovación e Universidades (MCNU), Agencia Estatal de Investigación (SAF2017‐83120‐C2‐1‐R and PID2020-113963RB-I00). Predoctoral grants/contracts from Gobierno de Aragon (IU-M and JP) are supported by ARAID Foundation. EG is funded by Ministerio de Ciencia, Innovación y Universidades (MCNU), and Agencia Estatal de Investigación (PID2020-113963RB-I00). MA and LS are funded by Postdoctoral Juan de la Cierva Contract. SR, LC, SH, and IU-M are funded by predoctoral contracts from Aragon Government. PL is funded by FPU predoctoral grants from Ministerio de Ciencia, Innovación e Universidades. Work at the Catalan Institute of Oncology is funded by the entity, the Instituto de Salud Carlos III and Ministerio de Economia y Competitividad, and co-funded by FEDER funds—a way to build Europe (PI20/00767), CIBERESP (grant CB07/02/2005), H2020 grant MoTriColor, and the Agency for Management of University and Research Grants (AGAUR) of the Catalan Government grant 2017SGR723. This work is supported by COST Action CA17118.
- Published
- 2022
46. Biological relevance of Granzymes A and K during E. coli sepsis
- Author
-
Ariel Ramirez-Labrada, Phillip I. Bird, Llipsy Santiago, Elena Tapia, Julián Pardo, Marcela Garzón-Tituaña, Maykel Arias, Iratxe Uranga-Murillo, Francisco J Roig, Patricia Perez Esteban, Eva M. Galvez, Cecilia Pesini, Ministerio de Ciencia, Innovación y Universidades (España), European Commission, Gobierno de Aragón, Asociación Española Contra el Cáncer, ARAID Foundation, Agencia Estatal de Investigación (España), Uranga Murillo, Iratxe, Tapia, Elena, Garzón, Marcela, Ramírez-Labrada, Ariel, Santiago, Llipsy, Pesini, Cecilia, Esteban, Patricia, Roig, Francisco J., Gálvez Buerba, Eva Mª, Bird, Phillip I., Pardo, Julián, Arias, Maykel, Uranga Murillo, Iratxe [0000-0001-8411-984X], Tapia, Elena [0000-0002-4275-1406], Garzón, Marcela [0000-0001-6778-0636], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Santiago, Llipsy [0000-0002-1861-5981], Pesini, Cecilia [0000-0002-8707-2722], Esteban, Patricia [0000-0003-4123-3524], Roig, Francisco J. [0000-0002-8853-2428], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Bird, Phillip I. [0000-0002-6695-606X], Pardo, Julián [0000-0003-0154-0730], and Arias, Maykel [0000-0002-9730-2210]
- Subjects
Inflammation ,Proteases ,Granzyme A ,Medicine (miscellaneous) ,Bacterial sepsis ,Biology ,medicine.disease ,Microbiology ,Proinflammatory cytokine ,Sepsis ,Granzyme ,medicine ,biology.protein ,Tumor necrosis factor alpha ,Granzyme K ,medicine.symptom ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) - Abstract
6 figures.-- Supplemenatry material available.--This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions., [Aims]: Recent in vitro findings suggest that the serine protease Granzyme K (GzmK) may act as a proinflammatory mediator. However, its role in sepsis is unknown. Here we aim to understand the role of GzmK in a mouse model of bacterial sepsis and compare it to the biological relevance of Granzyme A (GzmA)., [Methods]: Sepsis was induced in WT, GzmA-/- and GzmK-/- mice by an intraperitoneal injection of 2x108 CFU from E. coli. Mouse survival was monitored during 5 days. Levels of IL-1α, IL-1β, TNFα and IL-6 in plasma were measured and bacterial load in blood, liver and spleen was analyzed. Finally, profile of cellular expression of GzmA and GzmK was analyzed by FACS., [Results]: GzmA and GzmK are not involved in the control of bacterial infection. However, GzmA and GzmK deficient mice showed a lower sepsis score in comparison with WT mice, although only GzmA deficient mice exhibited increased survival. GzmA deficient mice also showed reduced expression of some proinflammatory cytokines like IL1-α, IL-β and IL-6. A similar result was found when extracellular GzmA was therapeutically inhibited in WT mice using serpinb6b, which improved survival and reduced IL-6 expression. Mechanistically, active extracellular GzmA induces the production of IL-6 in macrophages by a mechanism dependent on TLR4 and MyD88., [Conclusions]: These results suggest that although both proteases contribute to the clinical signs of E. coli-induced sepsis, inhibition of GzmA is sufficient to reduce inflammation and improve survival irrespectively of the presence of other inflammatory granzymes, like GzmK., This work was supported by grant SAF2017-83120-C2-1-R and PID2020-113963RBI00 from the Ministry of Science, Innovation and Universities and FEDER (Group B29_17R, Aragon Government). IU-M , MG-T and CP were supported by a PhD fellowships from Aragon Government, Ministry of Science, Innovation and Universities (FPI) and Asociacion Española contra el Cancer (AECC). MA and LS were supported by a post-doctoral fellowship “Juan de la Cierva-formación” (MA, LS) and "Juan de la Cierva-incorporacion" (MA) from the Ministry of Science, Innovation and Universities. JP was supported by ARAID Foundation.
- Published
- 2021
47. Granzyme A inhibition reduces inflammation and increases survival during abdominal sepsis
- Author
-
Julián Pardo, Pilar Luque, Ludovic Couty, Jose Luis Sierra-Monzón, Maykel Arias, Tatiana Khaliulina-Ushakova, José Ramón Paño-Pardo, Eric Camerer, Ariel Ramirez-Labrada, Cristina Seral, Phillip I. Bird, Laura Comas, Iratxe Uranga-Murillo, Marcela Garzón-Tituaña, Sonia Algarate, Elena Tapia, Llipsy Santiago, Eva M. Galvez, Elena Morte-Romea, Ministerio de Ciencia, Innovación y Universidades (España), European Commission, Gobierno de Aragón, ARAID Foundation, Garzón, Marcela, Sierra Monzón, José L., Comas, Laura, Santiago, Llipsy, Khaliulina, Tatiana, Uranga Murillo, Iratxe, Ramírez-Labrada, Ariel, Tapia, Elena, Morte Romea, Elena, Algarate, Sonia, Camerer, Eric, Bird, Phillip I., Luque Gómez, Pilar, Paño, José Ramón, Gálvez Buerba, Eva Mª, Pardo, Julián, Arias, Maykel, Garzón, Marcela [0000-0001-6778-0636], Sierra Monzón, José L. [0000-0002-8796-2717], Comas, Laura [0000-0002-3843-1231], Santiago, Llipsy [0000-0002-1861-5981], Khaliulina, Tatiana [0000-0002-7930-2129], Uranga Murillo, Iratxe [0000-0001-8411-984X], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Tapia, Elena [0000-0002-4275-1406], Morte Romea, Elena [0000-0001-9262-2461], Algarate, Sonia [0000-0002-0036-663], Camerer, Eric [0000-0002-6271-7125], Bird, Phillip I. [0000-0002-6695-606X], Luque Gómez, Pilar [0000-0001-7790-409X], Paño, José Ramón [0000-0002-9600-8116], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Pardo, Julián [0000-0003-0154-0730], and Arias, Maykel [0000-0002-9730-2210]
- Subjects
0301 basic medicine ,Male ,Medicine (miscellaneous) ,Granzymes ,Pathogenesis ,Mice ,0302 clinical medicine ,Molecular Targeted Therapy ,Precision Medicine ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Aged, 80 and over ,Mice, Knockout ,cecal ligation and puncture ,Granzyme A ,Middle Aged ,Killer Cells, Natural ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cytokines ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,Inflammation Mediators ,Research Paper ,Peritonitis ,Spleen ,Inflammation ,Proinflammatory cytokine ,Sepsis ,03 medical and health sciences ,medicine ,Extracellular ,Animals ,Humans ,Serpins ,Aged ,business.industry ,Interleukin-6 ,Macrophages ,Cecal ligation and puncture ,medicine.disease ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Disease Models, Animal ,030104 developmental biology ,Immunology ,business - Abstract
7 figures, 1 table., [Aims]: Peritonitis is one of the most common causes of sepsis, a serious syndrome characterized by a dysregulated systemic inflammatory response. Recent evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed by NK and T cells, could act as a proinflammatory mediator and could play an important role in the pathogenesis of sepsis. This work aims to analyze the role and the therapeutic potential of GzmA in the pathogenesis of peritoneal sepsis., [Methods]: The level of extracellular GzmA as well as GzmA activity were analyzed in serum from healthy volunteers and patients with confirmed peritonitis and were correlated with the Sequential Organ Failure Assessment (SOFA) score. Peritonitis was induced in C57Bl/6 (WT) and GzmA-/- mice by cecal ligation and puncture (CLP). Mice were treated intraperitoneally with antibiotics alone or in combination serpinb6b, a specific GzmA inhibitor, for 5 days. Mouse survival was monitored during 14 days, levels of some proinflammatory cytokines were measured in serum and bacterial load and diversity was analyzed in blood and spleen at different times., [Results]: Clinically, elevated GzmA was observed in serum from patients with abdominal sepsis suggesting that GzmA plays an important role in this pathology. In the CLP model GzmA deficient mice, or WT mice treated with an extracellular GzmA inhibitor, showed increased survival, which correlated with a reduction in proinflammatory markers in both serum and peritoneal lavage fluid. GzmA deficiency did not influence bacterial load in blood and spleen and GzmA did not affect bacterial replication in macrophages in vitro, indicating that GzmA has no role in bacterial control. Analysis of GzmA in lymphoid cells following CLP showed that it was mainly expressed by NK cells. Mechanistically, we found that extracellular active GzmA acts as a proinflammatory mediator in macrophages by inducing the TLR4-dependent expression of IL-6 and TNFα., [Conclusions]: Our findings implicate GzmA as a key regulator of the inflammatory response during abdominal sepsis and provide solid evidences about its therapeutic potential for the treatment of this severe pathology., This work was supported by grant SAF2017-83120-C2-1-R and SAF2014-54763-C2-2-R from the Ministry of Science, Innovation and Universities and FEDER (Group B29_17R, Aragon Government). MG and LS were supported by a PhD fellowship (FPI) from the Ministry of Science, Innovation and Universities. IUM was supported by a PhD fellowship from Aragon Government, MA was supported by a post-doctoral fellowship “Juan de la Cierva-formación” from the Ministry of Science, Innovation and Universities. JP was supported by ARAID Foundation.
- Published
- 2021
48. Integrated analysis of circulating immune cellular and soluble mediators reveals specific COVID19 signatures at hospital admission with utility for prediction of clinical outcomes
- Author
-
José Ramón Paño-Pardo, Sandra García-Mulero, Ariel Ramirez-Labrada, Borja Gracia-Tello, Rebeca Sanz-Pamplona, Luis Martínez-Lostao, Eva M. Galvez, Jose L. Sierra, Diego de Miguel, Cecilia Pesini, Maria del Mar Encabo-Berzosa, Elena Morte, Iratxe Uranga-Murillo, Llipsy Santiago, Julián Pardo, Sandra Hidalgo, Maykel Arias, European Commission, Gobierno de Aragón, Instituto de Salud Carlos III, Fundación Banco Santander, Universidad de Zaragoza, Agencia Estatal de Investigación (España), Fundación Inocente Inocente, Aspanoa, Asociación Carrera de la Mujer Ciudad de Monzón, Asociación Española Contra el Cáncer, Ministerio de Ciencia, Innovación y Universidades (España), ARAID Foundation, Uranga Murillo, Iratxe, Morte Romea, Elena, Hidalgo, Sandra, Pesini, Cecilia, García-Mulero, Sandra, Sierra Monzón, José L., Santiago, Llipsy, Arias, Maykel, Miguel, Diego de, Encabo-Berzosa, M. Mar, Gracia Tello, Borja, Sanz-Pamplona, Rebeca, Martínez Lostao, Luis, Gálvez Buerba, Eva Mª, Paño, José Ramón, Ramírez-Labrada, Ariel, Pardo, Julián, Uranga Murillo, Iratxe [0000-0001-8411-984X], Morte Romea, Elena [0000-0001-9262-2461], Hidalgo, Sandra [0000-0003-1629-9978], Pesini, Cecilia [0000-0002-8707-2722], García-Mulero, Sandra [0000-0003-4931-1267], Sierra Monzón, José L. [0000-0002-8796-2717], Santiago, Llipsy [0000-0002-1861-5981], Arias, Maykel [0000-0002-9730-2210], Miguel, Diego de [0000-0002-8486-8514], Encabo-Berzosa, M. Mar [0000-0001-5533-804X], Gracia Tello, Borja [0000-0003-3248-2908], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], Martínez Lostao, Luis [0000-0003-3043-147X], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Paño, José Ramón [0000-0002-9600-8116], Ramírez-Labrada, Ariel [0000-0002-3888-7036], and Pardo, Julián [0000-0003-0154-0730]
- Subjects
Adult ,Male ,Chemokine ,LAG3 ,COVID19 ,Medicine (miscellaneous) ,Inflammation ,NK cells ,CD8-Positive T-Lymphocytes ,CCL8 ,Severity of Illness Index ,Monocytes ,Proinflammatory cytokine ,Immune system ,Immunitat cel·lular ,medicine ,ULBP ,Humans ,Prospective Studies ,MIC ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Respiratory Tract Infections ,Aged ,Aged, 80 and over ,biology ,business.industry ,GzmA ,GzmB ,CXCL10 ,COVID-19 ,Middle Aged ,Immune checkpoint ,Cellular immunity ,Hospitalization ,Killer Cells, Natural ,Logistic Models ,Case-Control Studies ,CXCL9 ,Immunology ,biology.protein ,Cytokines ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,Chemokines ,business ,Biomarkers ,Research Paper - Abstract
6 figures, 6 tables.-- Supplementary material available., Coronavirus disease 2019 (COVID19), caused by SARS-CoV-2, is a complex disease, with a variety of clinical manifestations ranging from asymptomatic infection or mild cold-like symptoms to more severe cases requiring hospitalization and critical care. The most severe presentations seem to be related with a delayed, deregulated immune response leading to exacerbated inflammation and organ damage with close similarities to sepsis., [Methods]: In order to improve the understanding on the relation between host immune response and disease course, we have studied the differences in the cellular (monocytes, CD8+ T and NK cells) and soluble (cytokines, chemokines and immunoregulatory ligands) immune response in blood between Healthy Donors (HD), COVID19 and a group of patients with non-COVID19 respiratory tract infections (NON-COV-RTI). In addition, the immune response profile has been analyzed in COVID19 patients according to disease severity., [Results]: In comparison to HDs and patients with NON-COV-RTI, COVID19 patients show a heterogeneous immune response with the presence of both activated and exhausted CD8+ T and NK cells characterised by the expression of the immune checkpoint LAG3 and the presence of the adaptive NK cell subset. An increased frequency of adaptive NK cells and a reduction of NK cells expressing the activating receptors NKp30 and NKp46 correlated with disease severity. Although both activated and exhausted NK cells expressing LAG3 were increased in moderate/severe cases, unsupervised cell clustering analyses revealed a more complex scenario with single NK cells expressing more than one immune checkpoint (PD1, TIM3 and/or LAG3). A general increased level of inflammatory cytokines and chemokines was found in COVID19 patients, some of which like IL18, IL1RA, IL36B and IL31, IL2, IFNα and TNFα, CXCL10, CCL2 and CCL8 were able to differentiate between COVID19 and NON-COV-RTI and correlated with bad prognosis (IL2, TNFα, IL1RA, CCL2, CXCL10 and CXCL9). Notably, we found that soluble NKG2D ligands from the MIC and ULBPs families were increased in COVID19 compared to NON-COV-RTI and correlated with disease severity., [Conclusions]: Our results provide a detailed comprehensive analysis of the presence of activated and exhausted CD8+T, NK and monocyte cell subsets as well as extracellular inflammatory factors beyond cytokines/chemokines, specifically associated to COVID19. Importantly, multivariate analysis including clinical, demographical and immunological experimental variables have allowed us to reveal specific immune signatures to i) differentiate COVID19 from other infections and ii) predict disease severity and the risk of death., The authors would like to thank the Biobank of the Aragon Health System integrated in the Spanish National Biobanks Network and the Servicios Científico Técnicos de Citometria de Flujo del CIBA for their collaboration. Work in the JP laboratory is funded by the FEDER (Fondo Europeo de Desarrollo Regional, Gobierno de Aragón, Group B29_17R), Health National Institute Carlos III (COV20-00308), Aragón Government (Fondo COVID-19), Fundación Santander-Universidad de Zaragoza (Programa COVID-19), Agencia Estatal de Investigación (SAF2017-83120-C2-1-R; PID2020-113963RBI00), Fundación Inocente, ASPANOA and Carrera de la Mujer de Monzón. EMG is funded by Agencia Estatal de Investigación (SAF2017-83120-C2-1-R and PID2020-113963RB-I00). IUM and SH are supported by a PhD fellowship from Aragon Government, CP by a PhD fellowship from AECC, LS by a PhD fellowship (FPI) from the Ministry of Science, Innovation and Universities. DDM is supported by a postdoctoral fellowship 'Sara Borrell', and MA is supported by a postdoctoral fellowship 'Juan de la Cierva-incorporacion' from the Ministry of Science, Innovation and Universities. EM and BGT are supported by Rio Hortega contract. JP is supported by the ARAID Foundation.
- Published
- 2021
49. Cell death induced by cytotoxic CD8+ T cells is immunogenic and primes caspase-3-dependent spread immunity against endogenous tumor antigens
- Author
-
David Sancho, Paula Jaime-Sánchez, Nacho Aguilo, Sofía C. Khouili, Julián Pardo, Iratxe Uranga-Murillo, Maykel Arias, Asociación de Padres de Niños Oncológicos de Aragón, Gobierno de Aragón (España), Unión Europea. Fondo Europeo de Desarrollo Regional (FEDER/ERDF), Ministerio de Ciencia y Universidades (España), ARAID Foundation, Instituto de Salud Carlos III, Fundación ProCNIC, European Research Council, Universidad de Zaragoza, Jaime Sánchez, Paula, Uranga Murillo, Iratxe, Aguiló, Nacho, Chayeb Khouili, Sofia, Arias, Maykel, Sancho, David, Pardo, Julián, Jaime Sánchez, Paula [0000-0002-8731-4269, Uranga Murillo, Iratxe [0000-0001-8411-984X], Aguiló, Nacho [0000-0001-7897-9173], Chayeb Khouili, Sofia [0000-0002-7333-789X], Arias, Maykel [0000-0002-9730-2210], Sancho, David [0000-0003-2890-3984], and Pardo, Julián [0000-0003-0154-0730]
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Cytotoxic CD8+ T cells ,T-Lymphocytes ,Cytotoxicity ,Immunology ,chemical and pharmacologic phenomena ,Dendritic cells ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,Antigen ,Immunologic ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Antigens ,RC254-282 ,Pharmacology ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,Immunogenicity ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Caspases ,Cancer cell ,Cancer research ,Tumor immunology ,Molecular Medicine ,Immunogenic cell death ,Vaccine ,CD8 - Abstract
6 Figures, [Background] Elimination of cancer cells by some stimuli like chemotherapy and radiotherapy activates anticancer immunity after the generation of damage‐associated molecular patterns, a process recently named immunogenic cell death (ICD). Despite the recent advances in cancer immunotherapy, very little is known about the immunological consequences of cell death activated by cytotoxic CD8+ T (Tc) cells on cancer cells, that is, if Tc cells induce ICD on cancer cells and the molecular mechanisms involved., [Methods] ICD induced by Tc cells on EL4 cells was analyzed in tumor by vaccinating mice with EL4 cells killed in vitro or in vivo by Ag-specific Tc cells. EL4 cells and mutants thereof overexpressing Bcl-XL or a dominant negative mutant of caspase-3 and wild-type mice, as well as mice depleted of Tc cells and mice deficient in perforin, TLR4 and BATF3 were used. Ex vivo cytotoxicity of spleen cells from immunized mice was analyzed by flow cytometry. Expression of ICD signals (calreticulin, HMGB1 and interleukin (IL)-1β) was analyzed by flow cytometry and ELISA., [Results] Mice immunized with EL4.gp33 cells killed in vitro or in vivo by gp33-specific Tc cells were protected from parental EL4 tumor development. This result was confirmed in vivo by using ovalbumin (OVA) as another surrogate antigen. Perforin and TLR4 and BATF3-dependent type 1 conventional dendritic cells (cDC1s) were required for protection against tumor development, indicating cross-priming of Tc cells against endogenous EL4 tumor antigens. Tc cells induced ICD signals in EL4 cells. Notably, ICD of EL4 cells was dependent on caspase-3 activity, with reduced antitumor immunity generated by caspase-3–deficient EL4 cells. In contrast, overexpression of Bcl-XL in EL4 cells had no effect on induction of Tc cell antitumor response and protection., [Conclusions] Elimination of tumor cells by Ag-specific Tc cells is immunogenic and protects against tumor development by generating new Tc cells against EL4 endogenous antigens. This finding helps to explain the enhanced efficacy of T cell-dependent immunotherapy and provide a molecular basis to explain the epitope spread phenomenon observed during vaccination and chimeric antigen receptor (CAR)-T cell therapy. In addition, they suggest that caspase-3 activity in the tumor may be used as a biomarker to predict cancer recurrence during T cell-dependent immunotherapies., The authors would like to acknowledge the use of Servicios Científico Técnicos del CIBA (IACS-Universidad de Zaragoza) and Servicios Apoyo Investigación de la Universidad de Zaragoza.
- Published
- 2020
50. Extracellular Granzyme A Promotes Colorectal Cancer Development by Enhancing Gut Inflammation
- Author
-
Paula Jaime-Sánchez, Rosa del Campo, Eric Camerer, Luis Martínez-Lostao, Phillip I. Bird, Pilar M. Lanuza, Marcela Garzón, Gabriel Gil-Gómez, Pablo Pelegrín, Marta Castro, Anabel Alcalde, Angel Ferrandez, Julián Pardo, Llipsy Santiago, Eva M. Galvez, Guillermo Muñoz, Victor Moreno, Elena Tapia, Sunil S. Metkar, Elena Layunta, Marta Garrido, Maykel Arias, Raúl Peña, J. A. Uranga, Rebeca Sanz-Pamplona, Iratxe Uranga-Murillo, Ariel Ramirez-Labrada, Laura Comas, European Commission, Gobierno de Aragón, Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, Fundación Banco Santander, Universidad de Zaragoza, Ministerio de Educación, Cultura y Deporte (España), Ministerio de Empleo y Seguridad Social (España), Ministerio de Ciencia, Innovación y Universidades (España), ARAID Foundation, Santiago, Llipsy [0000-0002-1861-5981], Castro, Marta [0000-0001-8584-3979], Sanz-Pamplona, Rebeca [0000-0002-2187-3527], Ramírez-Labrada, Ariel [0000-0002-3888-7036], Moreno, Víctor [0000-0002-2818-5487], Layunta, Elena [0000-0001-5573-6144], Gil-Gómez, Gabriel [0000-0002-9790-7308], Peña, Raúl [0000-0002-1650-9720], Lanuza, Pilar M. [0000-0001-7328-2094], Comas, Laura [0000-0002-3843-1231], Jaime Sánchez, Paula [0000-0002-8731-4269], Uranga Murillo, Iratxe [0000-0001-8411-984X], Campo, Rosa del [0000-0003-1147-7923], Pelegrín, Pablo [0000-0002-9688-1804], Camerer, Eric [0000-0002-6271-7125], Martínez Lostao, Luis [0000-0003-3043-147X], Muñoz, Guillermo [0000-0001-8567-4327], Uranga, José A. [0000-0003-4656-8569], Alcalde, Anabel [0000-0002-9935-927X], Gálvez Buerba, Eva Mª [0000-0001-6928-5516], Ferrández, Ángel [0000-0003-2280-9372], Arias, Maykel [0000-0002-9730-2210], Pardo, Julián [0000-0003-0154-0730], Santiago, Llipsy, Castro, Marta, Sanz-Pamplona, Rebeca, Ramírez-Labrada, Ariel, Moreno, Víctor, Layunta, Elena, Gil-Gómez, Gabriel, Peña, Raúl, Lanuza, Pilar M., Comas, Laura, Jaime Sánchez, Paula, Uranga Murillo, Iratxe, Campo, Rosa del, Pelegrín, Pablo, Camerer, Eric, Martínez Lostao, Luis, Muñoz, Guillermo, Uranga, José A., Alcalde, Anabel, Gálvez Buerba, Eva Mª, Ferrández, Ángel, Arias, Maykel, and Pardo, Julián
- Subjects
0301 basic medicine ,Macrophage ,Carcinogenesis ,Inflammasomes ,Extracellular ,medicine.disease_cause ,Granzymes ,STAT3 ,chemistry.chemical_compound ,0302 clinical medicine ,Carcinogènesi ,Gut ,Mice, Knockout ,biology ,Dextran Sulfate ,NF-kappa B ,Acute Disease ,Disease Progression ,Cytokines ,medicine.symptom ,Inflammation Mediators ,Colorectal Neoplasms ,Colon ,Azoxymethane ,Inflammation ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,03 medical and health sciences ,Immune system ,Càncer colorectal ,medicine ,Animals ,Humans ,RNA, Messenger ,Granzyme ,business.industry ,Interleukin-6 ,Cancer ,medicine.disease ,Colorectal cancer ,digestive system diseases ,IL6 ,030104 developmental biology ,chemistry ,Cyclooxygenase 2 ,Chronic Disease ,Cancer research ,Granzyme A ,biology.protein ,business ,Extracellular Space ,030217 neurology & neurosurgery - Abstract
8 figures, 1 table.-- Electronic supplementary information available, If not properly regulated, the inflammatory immune response can promote carcinogenesis, as evident in colorectal cancer (CRC). Aiming to gain mechanistic insight into the link between inflammation and CRC, we perform transcriptomics analysis of human CRC, identifying a strong correlation between expression of the serine protease granzyme A (GzmA) and inflammation. In a dextran sodium sulfate and azoxymethane (DSS/AOM) mouse model, deficiency and pharmacological inhibition of extracellular GzmA both attenuate gut inflammation and prevent CRC development, including the initial steps of cell transformation and epithelial-to-mesenchymal transition. Mechanistically, extracellular GzmA induces NF-κB-dependent IL-6 production in macrophages, which in turn promotes STAT3 activation in cultured CRC cells. Accordingly, colon tissues from DSS/AOM-treated, GzmA-deficient animals present reduced levels of pSTAT3. By identifying GzmA as a proinflammatory protease that promotes CRC development, these findings provide information on mechanisms that link immune cell infiltration to cancer progression and present GzmA as a therapeutic target for CRC., This work was supported in part by FEDER/Gobierno de Aragón (group B29), Ministerio de Economia y Competitividad (SAF2014-54763-C2-1 and SAF2017-83120-C2-1-R to J.P. and SAF2014-54763-C2-2-R to E.M.G.), and Instituto de Salud Carlos III (PI13/00864 to L.M.-L.). Predoctoral grants/contracts from Fundacion Santander/Universidad de Zaragoza (to L.S. and M.A.A.), Gobierno de Aragon (to I.U.-M., L.C., L.S., and P.J.-S.), and FPU/Ministerio de Educación, Cultura y Deportes (to P.M.L.). I.U.-M. was supported by Fondo Garantía Empleo Juvenil/INAEM. M.A.A. has a Juan de la Cierva contract (Ministerio de Ciencia, Innovación y Universidades). J.P. was supported by Fundación Aragon I+D (ARAID).
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.