40 results on '"Fernandez-Irigoyen J"'
Search Results
2. P3.03I.05 DSTYK Inhibition as a Novel Strategy for Taxane-Based Chemotherapy Sensitation in Early and Advanced Lung Cancer
- Author
-
Echepare, M., Picabea, B., Arricibita, A., Teijeira, A., Pasquier, A., Zandueta, C., Santamaría, E., Fernández-Irigoyen, J., Romero, O., Sánchez-Céspedes, M., Lecanda, F., Hernández, J., Felip, E., Cruz, A., Provencio, M., Montuenga, L.M., and Valencia, K.
- Published
- 2024
- Full Text
- View/download PDF
3. 4P CBL-B inhibition overcomes PD-1/LAG-3 mediated resistance in lung cancer
- Author
-
Chocarro, L.D.E., Blanco, E., Fenández-Rubio, L., Garnica, M., Zuazo, M., Granda, M.J. Garcia, Bocanegra, A., Echaide, M., Johnston, C., Edwards, C.J., Legg, J., Pierce, A., Arasanz, H., Fernandez-Hinojal, G., Vera, R., Ausin, K., Santamaría, E., Fernández-Irigoyen, J., Kochan, G., and Escors, D.
- Published
- 2024
- Full Text
- View/download PDF
4. 15P CBL E3 ubiquitin ligases are key inhibitory regulators in PD-1/LAG-3 co-signaling in human cancers, targeted through bispecific co-blockade
- Author
-
Chocarro, L., Fernández-Rubio, L., Garcia Granda, M.J., Blanco Palmeiro, E., Bocanegra Gondan, A.I., Echaide, M., Garnica, M., Zuazo, M., Johnston, C., Edwards, C.J., Leggs, J., Pierce, A., Arasanz, H., Vera, R., Ausin, K., Santamaría, E., Fernández-Irigoyen, J., Kochan, G., and Escors, D.
- Published
- 2023
- Full Text
- View/download PDF
5. Soluble ST2 promotes oxidative stress and inflammation in cardiac fibroblasts: an in vitro and in vivo study in aortic stenosis
- Author
-
Matilla, L, Ibarrola, J, Arrieta, V, Garcia-Pena, A, Martinez-Martinez, E, Sadaba, R, Alvarez, V, Navarro, A, Fernandez-Celis, A, Gainza, A, Santamaria, E, Fernandez-Irigoyen, J, Bayes-Genis, A, Rossignol, P, and Lopez-Andres, N
- Subjects
mitofusin-1 ,inflammation ,fibroblasts ,sST2 ,oxidative stress - Abstract
Background: Soluble ST2 (interleukin 1 receptor-like 1) (sST2) is involved in inflammatory diseases and increased in heart failure (HF). We herein investigated sST2 effects on oxidative stress and inflammation in human cardiac fibroblasts and its pathological role in human aortic stenosis (AS). Methods and results: Using proteomics and immunodetection approaches, we have identified that sST2 down-regulated mitofusin-1 (MFN-1), a protein involved in mitochondrial fusion, in human cardiac fibroblasts. In parallel, sST2 increased nitrotyrosine, protein oxidation and peroxide production. Moreover, sST2 enhanced the secretion of pro-inflammatory cytokines interleukin (IL)-6, IL-1 beta and monocyte chemoattractant protein-1 (CCL-2). Pharmacological inhibition of transcriptional factor nuclear factor kappa B (NF kappa B) restored MFN-1 levels and improved oxidative status and inflammation in cardiac fibroblasts. Mito-Tempo, a mitochondria-specific superoxide scavenger, as well as Resveratrol, a general antioxidant, attenuated oxidative stress and inflammation induced by sST2. In myocardial biopsies from 26 AS patients, sST2 up-regulation paralleled a decrease in MFN-1. Cardiac sST2 inversely correlated with MFN-1 levels and positively associated with IL-6 and CCL-2 in myocardial biopsies from AS patients. Conclusions: sST2 affected mitochondrial fusion in human cardiac fibroblasts, increasing oxidative stress production and inflammatory markers secretion. The blockade of NF kappa B or mitochondrial reactive oxygen species restored MFN-1 expression, improving oxidative stress status and reducing inflammatory markers secretion. In human AS, cardiac sST2 levels associated with oxidative stress and inflammation. The present study reveals a new pathogenic pathway by which sST2 promotes oxidative stress and inflammation contributing to cardiac damage.
- Published
- 2019
6. 1074P Peripheral low-density neutrophils identify a subset of NSCLC patients with high PD-L1 tumor expression that can benefit from chemo-immunotherapy
- Author
-
Arasanz, H., Bocanegra Gondan, A.I., Teijeira, L., Garnica, M., Morilla Ruiz, I., Chocarro, L., Martínez-Aguillo, M., Ausin, K., Blanco, E., Zuazo, M., Fernandez-Hinojal, G., Echaide, M., Fernández-Rubio, L., Pineiro-Hermida, S., Ramos, P., Fernández-Irigoyen, J., Kochan, G., Vera, R., and Escors, D.
- Published
- 2022
- Full Text
- View/download PDF
7. 40P Identification of a PD-1/LAG-3 co-expression profile in human tumor-infiltrating T cells uncovers a T cell dysfunctionality genetic regulated programme
- Author
-
Chocarro, L., Blanco, E., Arasanz, H., Fernández-Rubio, L., Bocanegra, A.I., Echaide, M., Garnica, M., Ramos, P., Piñeiro-Hermida, S., García-Granda, M.J., Vera, R., Santamaría, E., Fernández-Irigoyen, J., Kochan, G., and Escors, D.
- Published
- 2022
- Full Text
- View/download PDF
8. Isoform-specific AKT inhibition differentially affects cell functions in pancreatic adenocarcinoma
- Author
-
Arasanz, H., primary, Gato-Cañas, M., additional, Zuazo, M., additional, Ibáñez-Vea, M., additional, Fernández-Hinojal, G., additional, Fernandez-Irigoyen, J., additional, Santamaria, E., additional, Kochan, G., additional, Vera, R., additional, and Escors, D., additional
- Published
- 2018
- Full Text
- View/download PDF
9. New predictive/prognostic biomarkers (BKs) in potentially resectable pancreatic adenocarcinoma (PDAC)
- Author
-
González, I., Viudez, A., Goñi, S., Ghanem, I., Amat, I., Arrazubi, V., Pérez, J., Hernandez, I., Feliu, J., Vera, R., and Fernández-Irigoyen, J.
- Published
- 2020
- Full Text
- View/download PDF
10. 9MO C-Myc regulates the cancer stem-cell phenotype differentially induced by the individual silencing of AKT isoforms in pancreatic adenocarcinoma
- Author
-
Arasanz, H., Hernández, C., Zuazo, M., Chocarro, L., Bocanegra-Gondan, A., Fernández-Hinojal, G., Blanco, E., Santamaría, E., Fernández-Irigoyen, J., Rodríguez, C., Vera, R., Kochan, G., and Escors, D.
- Published
- 2020
- Full Text
- View/download PDF
11. 37P - Adaption of pancreatic cancer cells to AKT1 inhibition induces the acquisition of cancer stem-cell like phenotype through upregulation of mitochondrial functions
- Author
-
Arasanz, H., Zuazo, M., Santamaría, E., Bocanegra, A.I., Gato-Cañas, M., Fernández-Hinojal, G., Hernández-Saez, C., Fernández-Irigoyen, J., Santamaria, E., Rodríguez-Ortigosa, C., Kochan, G., Vera, R., and Escors, D.
- Published
- 2019
- Full Text
- View/download PDF
12. P-202 - Search for new predictive/prognostic biomarkers in potentially resectable pancreatic ductal adenocarcinoma
- Author
-
González Borja, I., Viudez, A., Goñi, S., Pérez Sanz, J., Santamaria, E., López-López, C., Pazo, R., Fonseca, P. Jimenez, Arévalo, S., Feliu, J., and Fernández Irigoyen, J.
- Published
- 2019
- Full Text
- View/download PDF
13. Overexpression of Mirna-506 in Human Cholangiocytes Causes Primary Biliary Cholangitis-Like Features including Mitochondrial Dysfunction and Increased Sensitivity to Apoptosis
- Author
-
Erice, O., primary, Munoz-Garrido, P., additional, Vaquero, J., additional, Perugorria, M.J., additional, Fernandez-Barrena, M.G., additional, Sáez, E., additional, Arbelaiz, A., additional, Santos-Laso, Á., additional, Jimenez-Agüero, R., additional, Fernandez-Irigoyen, J., additional, Santamaria, E., additional, Torrano, V., additional, Carracedo, A., additional, Marzioni, M., additional, Prieto, J., additional, Beuers, U., additional, Elferink, R.O., additional, LaRusso, N.F., additional, Bujanda, L., additional, Marin, J.J.G., additional, and Banales, J.M., additional
- Published
- 2016
- Full Text
- View/download PDF
14. 26P - Isoform-specific AKT inhibition differentially affects cell functions in pancreatic adenocarcinoma
- Author
-
Arasanz, H., Gato-Cañas, M., Zuazo, M., Ibáñez-Vea, M., Fernández-Hinojal, G., Fernandez-Irigoyen, J., Santamaria, E., Kochan, G., Vera, R., and Escors, D.
- Published
- 2018
- Full Text
- View/download PDF
15. Proteomic analysis of human hepatoma cells expressing methionine adenosyltransferase I/III Characterization of DDX3X as a target of S-adenosylmethionine
- Author
-
Schröder, P.C. (Paul C.), Fernandez-Irigoyen, J. (Joaquín), Bigaud, E. (Emilie), Serna, A. (Antonio), Hernandez-Alcoceba, R. (Rubén), Lu, S.C. (Shelly C.), Mato, J.M. (José María), Prieto, J. (Jesús), and Corrales, F.J. (Fernando José)
- Subjects
Proteomics ,S-adenosylmethionine ,Mass spectrometry ,Hepatocellular carcinoma ,DDX3X ,2D-DIGE - Abstract
Methionine adenosyltransferase I/III (MATI/III) synthesizes S-adenosylmethionine (SAM) in quiescent hepatocytes. Its activity is compromised in most liver diseases including liver cancer. Since SAM is a driver of hepatocytes fate we have studied the effect of re-expressing MAT1A in hepatoma Huh7 cells using proteomics. MAT1A expression leads to SAM levels close to those found in quiescent hepatocytes and induced apoptosis. Normalization of intracellular SAM induced alteration of 128 proteins identified by 2D-DIGE and gel-free methods, accounting for deregulation of central cellular functions including apoptosis, cell proliferation and survival. Human Dead-box protein 3 (DDX3X), a RNA helicase regulating RNA splicing, export, transcription and translation was down-regulated upon MAT1A expression. Our data support the regulation of DDX3X levels by SAM in a concentration and time dependent manner. Consistently, DDX3X arises as a primary target of SAM and a principal intermediate of its antitumoral effect. Based on the parallelism between SAM and DDX3X along the progression of liver disorders, and the results reported here, it is tempting to suggest that reduced SAM in the liver may lead to DDX3X up-regulation contributing to the pathogenic process and that replenishment of SAM might prove to have beneficial effects, at least in part by reducing DDX3X levels. This article is part of a Special Issue entitled: Clinical Proteomics.
- Published
- 2012
16. Enzymatic activity of methionine adenosyltransferase variants identified in patients with persistent hypermethioninemia
- Author
-
Fernandez-Irigoyen, J. (Joaquín), Santamaria, E. (Enrique), Chien, Y-H (Yin-Hsiu), Hwu, W.L. (Wuh-Liang), Korman, S.H. (Stanley H.), Faghfoury, H. (Hanna), Schulze, A. (Andreas), Hoganson, G.E. (George E.), Stabler, S.P. (Sally P.), Allen, R.H. (Robert H.), Wagner, C. (Conrad), Mudd, S.H. (S. Harvey), and Corrales, F.J. (Fernando José)
- Subjects
S-adenosylmethionine ,Tripolyphosphate ,Methionine adenosyltransferase ,Hypermethioninemia - Abstract
Methionine adenosyltransferases (MAT's) are central enzymes in living organisms that have been conserved with a high degree of homology among species. In the liver, MAT I and III, tetrameric and dimeric isoforms of the same catalytic subunit encoded by the gene MAT1A, account for the predominant portion of total body synthesis of S-adenosylmethionine (SAM), a versatile sulfonium ion-containing molecule involved in a variety of vital metabolic reactions and in the control of hepatocyte proliferation and differentiation. During the past 15years 28 MAT1A mutations have been described in patients with elevated plasma methionines, total homocysteines at most only moderately elevated, and normal levels of tyrosine and other aminoacids. In this study we describe functional analyses that determine the MAT and tripolyphosphatase (PPPase) activities of 18 MAT1A variants, six of them novel, and none of them previously assayed for activity. With the exception of G69S and Y92H, all recombinant proteins showed impairment (usually severe) of MAT activity. Tripolyphosphate (PPPi) hydrolysis was decreased only in some mutant proteins but, when it was decreased MAT activity was always also impaired.
- Published
- 2010
17. 33P - Isoform-specific functions in pancreatic adenocarcinoma
- Author
-
Arasanz, H., Zuazo-Ibarra, M., Gato-Cañas, M., Ibáñez-Vea, M., Fernández-Hinojal, G., Fernández-Irigoyen, J., Santamaria, E., Kochan, G., Vera, R., and Escors, D.
- Published
- 2017
- Full Text
- View/download PDF
18. SAT-365 - Overexpression of Mirna-506 in Human Cholangiocytes Causes Primary Biliary Cholangitis-Like Features including Mitochondrial Dysfunction and Increased Sensitivity to Apoptosis
- Author
-
Erice, O., Munoz-Garrido, P., Vaquero, J., Perugorria, M.J., Fernandez-Barrena, M.G., Sáez, E., Arbelaiz, A., Santos-Laso, Á., Jimenez-Agüero, R., Fernandez-Irigoyen, J., Santamaria, E., Torrano, V., Carracedo, A., Marzioni, M., Prieto, J., Beuers, U., Elferink, R.O., LaRusso, N.F., Bujanda, L., Marin, J.J.G., and Banales, J.M.
- Published
- 2016
- Full Text
- View/download PDF
19. Redox regulation of methylthioadenosine phosphorylase in liver cells: molecular mechanism and functional implications
- Author
-
Fernandez-Irigoyen, J. (Joaquín)
- Subjects
- Cysteine oxidation,, Inflammatio, Methylthioadenosine, Methylthioadenosine phosphorylase, Oxidative stress, Sulfenic acid
- Abstract
MTAP (5'-methylthioadenosine phosphorylase) catalyses the reversible phosphorolytic cleavage of methylthioadenosine leading to the production of methylthioribose-1-phosphate and adenine. Deficient MTAP activity has been correlated with human diseases including cirrhosis and hepatocellular carcinoma. In the present study we have investigated the regulation of MTAP by ROS (reactive oxygen species). The results of the present study support the inactivation of MTAP in the liver of bacterial LPS (lipopolysaccharide)-challenged mice as well as in HepG2 cells after exposure to t-butyl hydroperoxide. Reversible inactivation of purified MTAP by hydrogen peroxide results from a reduction of V(max) and involves the specific oxidation of Cys(136) and Cys(223) thiols to sulfenic acid that may be further stabilized to sulfenyl amide intermediates. Additionally, we found that Cys(145) and Cys(211) were disulfide bonded upon hydrogen peroxide exposure. However, this modification is not relevant to the mediation of the loss of MTAP activity as assessed by site-directed mutagenesis. Regulation of MTAP by ROS might participate in the redox regulation of the methionine catabolic pathway in the liver. Reduced MTA (5'-deoxy-5'-methylthioadenosine)-degrading activity may compensate for the deficient production of the precursor S-adenosylmethionine, allowing maintenance of intracellular MTA levels that may be critical to ensure cellular adaptation to physiopathological conditions such as inflammation.
- Published
- 2008
20. TDP-43 regulates LC3ylation in neural tissue through ATG4B cryptic splicing inhibition.
- Author
-
Torres P, Rico-Rios S, Ceron-Codorniu M, Santacreu-Vilaseca M, Seoane-Miraz D, Jad Y, Ayala V, Mariño G, Beltran M, Miralles MP, Andrés-Benito P, Fernandez-Irigoyen J, Santamaria E, López-Otín C, Soler RM, Povedano M, Ferrer I, Pamplona R, Wood MJA, Varela MA, and Portero-Otin M
- Subjects
- Animals, Humans, Mice, Male, Spinal Cord metabolism, Spinal Cord pathology, Autophagy physiology, Mice, Knockout, RNA Splicing genetics, Female, Mice, Transgenic, Motor Neurons metabolism, Motor Neurons pathology, Oligonucleotides, Antisense pharmacology, Autophagy-Related Proteins metabolism, Autophagy-Related Proteins genetics, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Microtubule-Associated Proteins metabolism, Microtubule-Associated Proteins genetics, Amyotrophic Lateral Sclerosis metabolism, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis pathology, Cysteine Endopeptidases metabolism, Cysteine Endopeptidases genetics
- Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron disease with a mean survival time of three years. The 97% of the cases have TDP-43 nuclear depletion and cytoplasmic aggregation in motor neurons. TDP-43 prevents non-conserved cryptic exon splicing in certain genes, maintaining transcript stability, including ATG4B, which is crucial for autophagosome maturation and Microtubule-associated proteins 1A/1B light chain 3B (LC3B) homeostasis. In ALS mice (G93A), Atg4b depletion worsens survival rates and autophagy function. For the first time, we observed an elevation of LC3ylation in the CNS of both ALS patients and atg4b
-/- mouse spinal cords. Furthermore, LC3ylation modulates the distribution of ATG3 across membrane compartments. Antisense oligonucleotides (ASOs) targeting cryptic exon restore ATG4B mRNA in TARDBP knockdown cells. We further developed multi-target ASOs targeting TDP-43 binding sequences for a broader effect. Importantly, our ASO based in peptide-PMO conjugates show brain distribution post-IV administration, offering a non-invasive ASO-based treatment avenue for neurodegenerative diseases., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
21. PD-1/LAG-3 co-signaling profiling uncovers CBL ubiquitin ligases as key immunotherapy targets.
- Author
-
Chocarro L, Blanco E, Fernandez-Rubio L, Garnica M, Zuazo M, Garcia MJ, Bocanegra A, Echaide M, Johnston C, Edwards CJ, Legg J, Pierce AJ, Arasanz H, Fernandez-Hinojal G, Vera R, Ausin K, Santamaria E, Fernandez-Irigoyen J, Kochan G, and Escors D
- Subjects
- Humans, Animals, Mice, Neoplasms therapy, Neoplasms drug therapy, Neoplasms immunology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Cell Line, Tumor, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors pharmacology, Lymphocyte Activation Gene 3 Protein, Programmed Cell Death 1 Receptor metabolism, Programmed Cell Death 1 Receptor antagonists & inhibitors, Proto-Oncogene Proteins c-cbl metabolism, Proto-Oncogene Proteins c-cbl genetics, Immunotherapy methods, Signal Transduction drug effects, Antigens, CD metabolism, Antigens, CD genetics
- Abstract
Many cancer patients do not benefit from PD-L1/PD-1 blockade immunotherapies. PD-1 and LAG-3 co-upregulation in T-cells is one of the major mechanisms of resistance by establishing a highly dysfunctional state in T-cells. To identify shared features associated to PD-1/LAG-3 dysfunctionality in human cancers and T-cells, multiomic expression profiles were obtained for all TCGA cancers immune infiltrates. A PD-1/LAG-3 dysfunctional signature was found which regulated immune, metabolic, genetic, and epigenetic pathways, but especially a reinforced negative regulation of the TCR signalosome. These results were validated in T-cell lines with constitutively active PD-1, LAG-3 pathways and their combination. A differential analysis of the proteome of PD-1/LAG-3 T-cells showed a specific enrichment in ubiquitin ligases participating in E3 ubiquitination pathways. PD-1/LAG-3 co-blockade inhibited CBL-B expression, while the use of a bispecific drug in clinical development also repressed C-CBL expression, which reverted T-cell dysfunctionality in lung cancer patients resistant to PD-L1/PD-1 blockade. The combination of CBL-B-specific small molecule inhibitors with anti-PD-1/anti-LAG-3 immunotherapies demonstrated notable therapeutic efficacy in models of lung cancer refractory to immunotherapies, overcoming PD-1/LAG-3 mediated resistance., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. Drug-Induced Reorganisation of Lipid Metabolism Limits the Therapeutic Efficacy of Ponatinib in Glioma Stem Cells.
- Author
-
Aldaz P, Olias-Arjona A, Lasheras-Otero I, Ausin K, Redondo-Muñoz M, Wellbrock C, Santamaria E, Fernandez-Irigoyen J, and Arozarena I
- Abstract
The standard of care for glioblastoma (GBM) involves surgery followed by adjuvant radio- and chemotherapy, but often within months, patients relapse, and this has been linked to glioma stem cells (GSCs), self-renewing cells with increased therapy resistance. The identification of the epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR) as key players in gliomagenesis inspired the development of inhibitors targeting these tyrosine kinases (TKIs). However, results from clinical trials testing TKIs have been disappointing, and while the role of GSCs in conventional therapy resistance has been extensively studied, less is known about resistance of GSCs to TKIs. In this study, we have used compartmentalised proteomics to analyse the adaptive response of GSCs to ponatinib, a TKI with activity against PDGFR. The analysis of differentially expressed proteins revealed that GSCs respond to ponatinib by broadly rewiring lipid metabolism, involving fatty acid beta-oxidation, cholesterol synthesis, and sphingolipid degradation. Inhibiting each of these metabolic pathways overcame ponatinib adaptation of GSCs, but interrogation of patient data revealed sphingolipid degradation as the most relevant pathway in GBM. Our data highlight that targeting lipid metabolism, and particularly sphingolipid degradation in combinatorial therapies, could improve the outcome of TKI therapies using ponatinib in GBM.
- Published
- 2024
- Full Text
- View/download PDF
23. Oleuropein-driven reprogramming of the myeloid cell compartment to sensitise tumours to PD-1/PD-L1 blockade strategies.
- Author
-
Blanco E, Silva-Pilipich N, Bocanegra A, Chocarro L, Procopio A, Ausín K, Fernandez-Irigoyen J, Fernández L, Razquin N, Igea A, Garnica M, Echaide M, Arasanz H, Vera R, Escors D, Smerdou C, and Kochan G
- Subjects
- Humans, Animals, Mice, Programmed Cell Death 1 Receptor, Immune Checkpoint Inhibitors pharmacology, Myeloid Cells, Immunotherapy, Tumor Microenvironment, B7-H1 Antigen, Lung Neoplasms drug therapy, Iridoid Glucosides
- Abstract
Background: Previous studies have shown that functional systemic immunity is required for the efficacy of PD-1/PD-L1 blockade immunotherapies in cancer. Hence, systemic reprogramming of immunosuppressive dysfunctional myeloid cells could overcome resistance to cancer immunotherapy., Methods: Reprogramming of tumour-associated myeloid cells with oleuropein was studied by quantitative differential proteomics, phenotypic and functional assays in mice and lung cancer patients. Combinations of oleuropein and two different delivery methods of anti-PD-1 antibodies were tested in colorectal cancer tumour models and in immunotherapy-resistant lung cancer models., Results: Oleuropein treatment reprogrammed monocytic and granulocytic myeloid-derived suppressor cells, and tumour-associated macrophages towards differentiation of immunostimulatory subsets. Oleuropein regulated major differentiation programmes associated to immune modulation in myeloid cells, which potentiated T cell responses and PD-1 blockade. PD-1 antibodies were delivered by two different strategies, either systemically or expressed within tumours using a self-amplifying RNA vector. Combination anti-PD-1 therapies with oleuropein increased tumour infiltration by immunostimulatory dendritic cells in draining lymph nodes, leading to systemic antitumour T cell responses. Potent therapeutic activities were achieved in colon cancer and lung cancer models resistant to immunotherapies, even leading to complete tumour regression., Discussion: Oleuropein significantly improves the outcome of PD-1/PD-L1 blockade immunotherapy strategies by reprogramming myeloid cells., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. Signature-driven repurposing of Midostaurin for combination with MEK1/2 and KRASG12C inhibitors in lung cancer.
- Author
-
Macaya I, Roman M, Welch C, Entrialgo-Cadierno R, Salmon M, Santos A, Feliu I, Kovalski J, Lopez I, Rodriguez-Remirez M, Palomino-Echeverria S, Lonfgren SM, Ferrero M, Calabuig S, Ludwig IA, Lara-Astiaso D, Jantus-Lewintre E, Guruceaga E, Narayanan S, Ponz-Sarvise M, Pineda-Lucena A, Lecanda F, Ruggero D, Khatri P, Santamaria E, Fernandez-Irigoyen J, Ferrer I, Paz-Ares L, Drosten M, Barbacid M, Gil-Bazo I, and Vicent S
- Subjects
- Humans, Drug Repositioning, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Drug Combinations, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Mutation, Cell Line, Tumor, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology
- Abstract
Drug combinations are key to circumvent resistance mechanisms compromising response to single anti-cancer targeted therapies. The implementation of combinatorial approaches involving MEK1/2 or KRASG12C inhibitors in the context of KRAS-mutated lung cancers focuses fundamentally on targeting KRAS proximal activators or effectors. However, the antitumor effect is highly determined by compensatory mechanisms arising in defined cell types or tumor subgroups. A potential strategy to find drug combinations targeting a larger fraction of KRAS-mutated lung cancers may capitalize on the common, distal gene expression output elicited by oncogenic KRAS. By integrating a signature-driven drug repurposing approach with a pairwise pharmacological screen, here we show synergistic drug combinations consisting of multi-tyrosine kinase PKC inhibitors together with MEK1/2 or KRASG12C inhibitors. Such combinations elicit a cytotoxic response in both in vitro and in vivo models, which in part involves inhibition of the PKC inhibitor target AURKB. Proteome profiling links dysregulation of MYC expression to the effect of both PKC inhibitor-based drug combinations. Furthermore, MYC overexpression appears as a resistance mechanism to MEK1/2 and KRASG12C inhibitors. Our study provides a rational framework for selecting drugs entering combinatorial strategies and unveils MEK1/2- and KRASG12C-based therapies for lung cancer., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
25. The Regulators of Peroxisomal Acyl-Carnitine Shuttle CROT and CRAT Promote Metastasis in Melanoma.
- Author
-
Lasheras-Otero I, Feliu I, Maillo A, Moreno H, Redondo-Muñoz M, Aldaz P, Bocanegra A, Olias-Arjona A, Lecanda F, Fernandez-Irigoyen J, Santamaria E, Larrayoz IM, Gomez-Cabrero D, Wellbrock C, Vicent S, and Arozarena I
- Subjects
- Mice, Animals, Carnitine O-Acetyltransferase genetics, Carnitine O-Acetyltransferase metabolism, Carnitine Acyltransferases genetics, Carnitine Acyltransferases metabolism, Ranolazine, Oxidation-Reduction, Fatty Acids metabolism, Carnitine metabolism, Neoplastic Cells, Circulating, Melanoma drug therapy
- Abstract
Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. New In Vivo Approach to Broaden the Thioredoxin Family Interactome in Chloroplasts.
- Author
-
Ancín M, Fernandez-Irigoyen J, Santamaria E, Larraya L, Fernández-San Millán A, Veramendi J, and Farran I
- Abstract
Post-translational redox modifications provide an important mechanism for the control of major cellular processes. Thioredoxins (Trxs), which are key actors in this regulatory mechanism, are ubiquitous proteins that catalyse thiol-disulfide exchange reactions. In chloroplasts, Trx f, Trx m and NADPH-dependent Trx reductase C (NTRC) have been identified as transmitters of the redox signal by transferring electrons to downstream target enzymes. The number of characterised Trx targets has greatly increased in the last few years, but most of them were determined using in vitro procedures lacking isoform specificity. With this background, we have developed a new in vivo approach based on the overexpression of His-tagged single-cysteine mutants of Trx f, Trx m or NTRC into Nicotiana benthamiana plants. The over-expressed mutated Trxs, capable of forming a stable mixed disulfide bond with target proteins in plants, were immobilised on affinity columns packed with Ni-NTA agarose, and the covalently linked targets were eluted with dithiothreitol and identified by mass spectrometry-based proteomics. The in vivo approach allowed identification of 6, 9 and 42 new potential targets for Trx f, Trx m and NTRC, respectively, and an apparent specificity between NTRC and Trxs was achieved. Functional analysis showed that these targets are involved in several cellular processes., Competing Interests: The authors declare no conflict of interest. The funding organisations had no role in the design of the study, the collection, analysis, or interpretation of the data, the writing of the manuscript, or the decision to publish the results.
- Published
- 2022
- Full Text
- View/download PDF
27. α-Synuclein molecular behavior and nigral proteomic profiling distinguish subtypes of Lewy body disorders.
- Author
-
Martinez-Valbuena I, Swinkin E, Santamaria E, Fernandez-Irigoyen J, Sackmann V, Kim A, Li J, Gonzalez-Latapi P, Kuhlman G, Bhowmick SS, Visanji NP, Lang AE, and Kovacs GG
- Subjects
- Disease Progression, Humans, Lewy Bodies metabolism, Lewy Bodies pathology, Proteomics methods, Lewy Body Disease metabolism, Lewy Body Disease pathology, Substantia Nigra metabolism, alpha-Synuclein metabolism
- Abstract
Lewy body disorders (LBD), characterized by the deposition of misfolded α-synuclein (α-Syn), are clinically heterogeneous. Although the distribution of α-Syn correlates with the predominant clinical features, the burden of pathology does not fully explain the observed variability in clinical presentation and rate of disease progression. We hypothesized that this heterogeneity might reflect α-Syn molecular diversity, between both patients and different brain regions. Using an ultra-sensitive assay, we evaluated α-Syn seeding in 8 brain regions from 30 LBD patients with different clinical phenotypes and disease durations. Comparing seeding across the clinical phenotypes revealed that hippocampal α-Syn from patients with a cognitive-predominant phenotype had significantly higher seeding capacity than that derived from patients with a motor-predominant phenotype, whose nigral-derived α-Syn in turn had higher seeding capacity than that from cognitive-predominant patients. Interestingly, α-Syn from patients with rapid disease progression (< 3 years to development of advanced disease) had the highest nigral seeding capacity of all the patients included. To validate these findings and explore factors underlying seeding heterogeneity, we performed in vitro toxicity assays, and detailed neuropathological and biochemical examinations. Furthermore, and for the first time, we performed a proteomic-wide profiling of the substantia nigra from 5 high seeder and 5 low seeder patients. The proteomic data suggests a significant disruption in mitochondrial function and lipid metabolism in high seeder cases compared to the low seeders. These observations suggest that distinct molecular populations of α-Syn may contribute to heterogeneity in phenotypes and progression rates in LBD and imply that effective therapeutic strategies might need to be directed at an ensemble of differently misfolded α-Syn species, with the relative contribution of their differing impacts accounting for heterogeneity in the neurodegenerative process., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
28. Multi-laboratory experiment PME11 for the standardization of phosphoproteome analysis.
- Author
-
Colomé N, Abian J, Aloria K, Arizmendi JM, Barceló-Batllori S, Braga-Lagache S, Burlet-Schiltz O, Carrascal M, Casal JI, Chicano-Gálvez E, Chiva C, Clemente LF, Elortza F, Estanyol JM, Fernandez-Irigoyen J, Fernández-Puente P, Fidalgo MJ, Froment C, Fuentes M, Fuentes-Almagro C, Gay M, Hainard A, Heller M, Hernández ML, Ibarrola N, Iloro I, Kieselbach T, Lario A, Locard-Paulet M, Marina-Ramírez A, Martín L, Morato-López E, Muñoz J, Navajas R, Odena MA, Odriozola L, de Oliveira E, Paradela A, Pasquarello C, de Los Rios V, Ruiz-Romero C, Sabidó E, Sánchez Del Pino M, Sancho J, Santamaría E, Schaeffer-Reiss C, Schneider J, de la Torre C, Valero ML, Vilaseca M, Wu S, Wu L, Ximénez de Embún P, Canals F, and Corrales FJ
- Subjects
- Laboratories, Phosphoproteins analysis, Phosphorylation, Reference Standards, Reproducibility of Results, Proteome analysis, Proteomics methods
- Abstract
Global analysis of protein phosphorylation by mass spectrometry proteomic techniques has emerged in the last decades as a powerful tool in biological and biomedical research. However, there are several factors that make the global study of the phosphoproteome more challenging than measuring non-modified proteins. The low stoichiometry of the phosphorylated species and the need to retrieve residue specific information require particular attention on sample preparation, data acquisition and processing to ensure reproducibility, qualitative and quantitative robustness and ample phosphoproteome coverage in phosphoproteomic workflows. Aiming to investigate the effect of different variables in the performance of proteome wide phosphoprotein analysis protocols, ProteoRed-ISCIII and EuPA launched the Proteomics Multicentric Experiment 11 (PME11). A reference sample consisting of a yeast protein extract spiked in with different amounts of a phosphomix standard (Sigma/Merck) was distributed to 31 laboratories around the globe. Thirty-six datasets from 23 laboratories were analyzed. Our results indicate the suitability of the PME11 reference sample to benchmark and optimize phosphoproteomics strategies, weighing the influence of different factors, as well as to rank intra and inter laboratory performance., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. A Proteomic Atlas of Lineage and Cancer-Polarized Expression Modules in Myeloid Cells Modeling Immunosuppressive Tumor-Infiltrating Subsets.
- Author
-
Blanco E, Ibañez-Vea M, Hernandez C, Drici L, Martínez de Morentin X, Gato M, Ausin K, Bocanegra A, Zuazo M, Chocarro L, Arasanz H, Fernandez-Hinojal G, Fernandez-Irigoyen J, Smerdou C, Garnica M, Echaide M, Fernandez L, Morente P, Ramos-Castellanos P, Llopiz D, Santamaria E, Larsen MR, Escors D, and Kochan G
- Abstract
Monocytic and granulocytic myeloid-derived suppressor cells together with tumor-infiltrating macrophages constitute the main tumor-infiltrating immunosuppressive myeloid populations. Due to the phenotypic resemblance to conventional myeloid cells, their identification and purification from within the tumors is technically difficult and makes their study a challenge. We differentiated myeloid cells modeling the three main tumor-infiltrating types together with uncommitted macrophages, using ex vivo differentiation methods resembling the tumor microenvironment. The phenotype and proteome of these cells was compared to identify linage-dependent relationships and cancer-specific interactome expression modules. The relationships between monocytic MDSCs and TAMs, monocytic MDSCs and granulocytic MDSCs, and hierarchical relationships of expression networks and transcription factors due to lineage and cancer polarization were mapped. Highly purified immunosuppressive myeloid cell populations that model tumor-infiltrating counterparts were systematically analyzed by quantitative proteomics. Full functional interactome maps have been generated to characterize at high resolution the relationships between the three main myeloid tumor-infiltrating cell types. Our data highlights the biological processes related to each cell type, and uncover novel shared and differential molecular targets. Moreover, the high numbers and fidelity of ex vivo-generated subsets to their natural tumor-shaped counterparts enable their use for validation of new treatments in high-throughput experiments.
- Published
- 2021
- Full Text
- View/download PDF
30. Soluble St2 Induces Cardiac Fibroblast Activation and Collagen Synthesis via Neuropilin-1.
- Author
-
Matilla L, Arrieta V, Jover E, Garcia-Peña A, Martinez-Martinez E, Sadaba R, Alvarez V, Navarro A, Fernandez-Celis A, Gainza A, Santamaria E, Fernandez-Irigoyen J, Rossignol P, Zannad F, and Lopez-Andres N
- Subjects
- Animals, Blotting, Western, CRISPR-Cas Systems, Enzyme-Linked Immunosorbent Assay, Male, NF-kappa B metabolism, Proteomics methods, Rats, Rats, Wistar, Real-Time Polymerase Chain Reaction, Collagen metabolism, Fibroblasts metabolism, Myocardium cytology, Myocardium metabolism, Neuropilin-1 metabolism, Receptors, Interleukin-1 metabolism
- Abstract
Circulating levels of soluble interleukin 1 receptor-like 1 (sST2) are increased in heart failure and associated with poor outcome, likely because of the activation of inflammation and fibrosis. We investigated the pathogenic role of sST2 as an inductor of cardiac fibroblasts activation and collagen synthesis. The effects of sST2 on human cardiac fibroblasts was assessed using proteomics and immunodetection approaches to evidence the upregulation of neuropilin-1 (NRP-1), a regulator of the profibrotic transforming growth factor (TGF)-β1. In parallel, sST2 increased fibroblast activation, collagen and fibrosis mediators. Pharmacological inhibition of nuclear factor-kappa B (NF-κB) restored NRP-1 levels and blocked profibrotic effects induced by sST2. In NRP-1 knockdown cells, sST2 failed to induce fibroblast activation and collagen synthesis. Exogenous NRP-1 enhanced cardiac fibroblast activation and collagen synthesis via NF-κB. In a pressure overload rat model, sST2 was elevated in association with cardiac fibrosis and was positively correlated with NRP-1 expression. Our study shows that sST2 induces human cardiac fibroblasts activation, as well as the synthesis of collagen and profibrotic molecules. These effects are mediated by NRP-1. The blockade of NF-κB restored NRP-1 expression, improving the profibrotic status induced by sST2. These results show a new pathogenic role for sST2 and its mediator, NRP-1, as cardiac fibroblast activators contributing to cardiac fibrosis.
- Published
- 2020
- Full Text
- View/download PDF
31. Neuroanatomical Quantitative Proteomics Reveals Common Pathogenic Biological Routes between Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD).
- Author
-
Iridoy MO, Zubiri I, Zelaya MV, Martinez L, Ausín K, Lachen-Montes M, Santamaría E, Fernandez-Irigoyen J, and Jericó I
- Subjects
- Amyotrophic Lateral Sclerosis pathology, Biomarkers, Female, Frontotemporal Dementia pathology, Humans, Male, Mitochondria metabolism, Motor Neurons metabolism, Organ Specificity, Prohibitins, Signal Transduction, Spinal Cord metabolism, Spinal Cord pathology, Amyotrophic Lateral Sclerosis metabolism, Frontotemporal Dementia metabolism, Proteome, Proteomics methods
- Abstract
(1) Background: Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders with an overlap in clinical presentation and neuropathology. Common and differential mechanisms leading to protein expression changes and neurodegeneration in ALS and FTD were studied trough a deep neuroproteome mapping of the spinal cord. (2) Methods: A liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of the spinal cord from ALS-TAR DNA-binding protein 43 ( TDP-43 ) subjects, ubiquitin-positive frontotemporal lobar degeneration (FTLD-U) subjects and controls without neurodegenerative disease was performed. (3) Results: 281 differentially expressed proteins were detected among ALS versus controls, while 52 proteins were dysregulated among FTLD-U versus controls. Thirty-three differential proteins were shared between both syndromes. The resulting data was subjected to network-driven proteomics analysis, revealing mitochondrial dysfunction and metabolic impairment, both for ALS and FTLD-U that could be validated through the confirmation of expression levels changes of the Prohibitin ( PHB ) complex. (4) Conclusions: ALS-TDP-43 and FTLD-U share molecular and functional alterations, although part of the proteostatic impairment is region- and disease-specific. We have confirmed the involvement of specific proteins previously associated with ALS (Galectin 2 ( LGALS3 ), Transthyretin ( TTR ), Protein S100-A6 ( S100A6 ), and Protein S100-A11 ( S100A11 )) and have shown the involvement of proteins not previously described in the ALS context (Methanethiol oxidase ( SELENBP1 ), Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 ( PIN-1 ), Calcyclin-binding protein ( CACYBP ) and Rho-associated protein kinase 2 ( ROCK2 )).
- Published
- 2018
- Full Text
- View/download PDF
32. Aldosterone Impairs Mitochondrial Function in Human Cardiac Fibroblasts via A-Kinase Anchor Protein 12.
- Author
-
Ibarrola J, Sadaba R, Martinez-Martinez E, Garcia-Peña A, Arrieta V, Alvarez V, Fernández-Celis A, Gainza A, Cachofeiro V, Santamaria E, Fernandez-Irigoyen J, Jaisser F, and Lopez-Andres N
- Subjects
- A Kinase Anchor Proteins antagonists & inhibitors, A Kinase Anchor Proteins metabolism, Aged, Aged, 80 and over, Aldosterone pharmacology, Animals, Aortic Valve Stenosis metabolism, Aortic Valve Stenosis pathology, Aortic Valve Stenosis surgery, CRISPR-Cas Systems, Case-Control Studies, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins metabolism, DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism, Female, Fibroblasts drug effects, Fibroblasts pathology, Gene Expression Regulation, Gene Knockdown Techniques, Humans, Male, Middle Aged, Mitochondria drug effects, Mitochondria metabolism, Mitochondria pathology, Myocardium pathology, Organelle Biogenesis, Oxidative Stress, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Rats, Rats, Wistar, Signal Transduction, A Kinase Anchor Proteins genetics, Aldosterone metabolism, Aortic Valve Stenosis genetics, Cell Cycle Proteins genetics, Fibroblasts metabolism, Myocardium metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics
- Abstract
Aldosterone (Aldo) contributes to mitochondrial dysfunction and cardiac oxidative stress. Using a proteomic approach, A-kinase anchor protein (AKAP)-12 has been identified as a down-regulated protein by Aldo in human cardiac fibroblasts. We aim to characterize whether AKAP-12 down-regulation could be a deleterious mechanism which induces mitochondrial dysfunction and oxidative stress in cardiac cells. Aldo down-regulated AKAP-12 via its mineralocorticoid receptor, increased oxidative stress and induced mitochondrial dysfunction characterized by decreased mitochondrial-DNA and Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expressions in human cardiac fibroblasts. CRISPR/Cas9-mediated knock-down of AKAP-12 produced similar deleterious effects in human cardiac fibroblasts. CRISPR/Cas9-mediated activation of AKAP-12 blunted Aldo effects on mitochondrial dysfunction and oxidative stress in human cardiac fibroblasts. In Aldo-salt-treated rats, cardiac AKAP-12, mitochondrial-DNA and PGC-1α expressions were decreased and paralleled increased oxidative stress. In myocardial biopsies from patients with aortic stenosis (AS, n = 26), AKAP-12, mitochondrial-DNA and PGC-1α expressions were decreased as compared to Controls (n = 13). Circulating Aldo levels inversely correlated with cardiac AKAP-12. PGC-1α positively associated with AKAP-12 and with mitochondrial-DNA. Aldo decreased AKAP-12 expression, impairing mitochondrial biogenesis and increasing cardiac oxidative stress. AKAP-12 down-regulation triggered by Aldo may represent an important event in the development of mitochondrial dysfunction and cardiac oxidative stress.
- Published
- 2018
- Full Text
- View/download PDF
33. MicroRNA-506 promotes primary biliary cholangitis-like features in cholangiocytes and immune activation.
- Author
-
Erice O, Munoz-Garrido P, Vaquero J, Perugorria MJ, Fernandez-Barrena MG, Saez E, Santos-Laso A, Arbelaiz A, Jimenez-Agüero R, Fernandez-Irigoyen J, Santamaria E, Torrano V, Carracedo A, Ananthanarayanan M, Marzioni M, Prieto J, Beuers U, Oude Elferink RP, LaRusso NF, Bujanda L, Marin JJG, and Banales JM
- Subjects
- Apoptosis, Bile Ducts, Intrahepatic metabolism, Cell Culture Techniques, Cell Migration Assays, Cell Proliferation, Cytokines metabolism, Fluorescent Antibody Technique, Gene Expression Regulation genetics, Humans, Immunoblotting, Mass Spectrometry, Oxidative Stress, Proteomics, Signal Transduction genetics, Bile Ducts, Intrahepatic pathology, Epithelial Cells metabolism, Liver Cirrhosis, Biliary metabolism, MicroRNAs metabolism
- Abstract
Primary biliary cholangitis (PBC) is a chronic cholestatic liver disease associated with autoimmune phenomena targeting intrahepatic bile duct cells (cholangiocytes). Although its etiopathogenesis remains obscure, development of antimitochondrial autoantibodies against pyruvate dehydrogenase complex E2 is a common feature. MicroRNA (miR) dysregulation occurs in liver and immune cells of PBC patients, but its functional relevance is largely unknown. We previously reported that miR-506 is overexpressed in PBC cholangiocytes and directly targets both Cl
- / HCO3- anion exchanger 2 and type III inositol 1,4,5-trisphosphate receptor, leading to cholestasis. Here, the regulation of miR-506 gene expression and its role in cholangiocyte pathophysiology and immune activation was studied. Several proinflammatory cytokines overexpressed in PBC livers (such as interleukin-8 [IL8], IL12, IL17, IL18, and tumor necrosis factor alpha) stimulated miR-506 promoter activity in human cholangiocytes, as revealed by luciferase reporter assays. Experimental overexpression of miR-506 in cholangiocytes dysregulated the cell proteomic profile (by mass spectrometry), affecting proteins involved in different biological processes including mitochondrial metabolism. In cholangiocytes, miR-506 (1) induced dedifferentiation with down-regulation of biliary and epithelial markers together with up-regulation of mesenchymal, proinflammatory, and profibrotic markers; (2) impaired cell proliferation and adhesion; (3) increased oxidative and endoplasmic reticulum stress; (4) caused DNA damage; and (5) sensitized to caspase-3-dependent apoptosis induced by cytotoxic bile acids. These events were also associated with impaired energy metabolism in mitochondria (proton leak and less adenosine triphosphate production) and pyruvate dehydrogenase complex E2 overexpression. Coculture of miR-506 overexpressing cholangiocytes with PBC immunocytes induced activation and proliferation of PBC immunocytes., Conclusion: Different proinflammatory cytokines enhance the expression of miR-506 in biliary epithelial cells; miR-506 induces PBC-like features in cholangiocytes and promotes immune activation, representing a potential therapeutic target for PBC patients. (Hepatology 2018;67:1420-1440)., (© 2017 by the American Association for the Study of Liver Diseases.)- Published
- 2018
- Full Text
- View/download PDF
34. A multicentric study to evaluate the use of relative retention times in targeted proteomics.
- Author
-
Vialas V, Colomé-Calls N, Abian J, Aloria K, Alvarez-Llamas G, Antúnez O, Arizmendi JM, Azkargorta M, Barceló-Batllori S, Barderas MG, Blanco F, Casal JI, Casas V, de la Torre C, Chicano-Gálvez E, Elortza F, Espadas G, Estanyol JM, Fernandez-Irigoyen J, Fernandez-Puente P, Fidalgo MJ, Fuentes M, Gay M, Gil C, Hainard A, Hernaez ML, Ibarrola N, Kopylov AT, Lario A, Lopez JA, López-Lucendo M, Marcilla M, Marina-Ramírez A, Marko-Varga G, Martín L, Mora MI, Morato-López E, Muñoz J, Odena MA, de Oliveira E, Orera I, Ortea I, Pasquarello C, Ray KB, Rezeli M, Ruppen I, Sabidó E, Del Pino MMS, Sancho J, Santamaría E, Vazquez J, Vilaseca M, Vivanco F, Walters JJ, Zgoda VG, Corrales FJ, Canals F, and Paradela A
- Subjects
- Biomedical Research standards, Chromatography, Liquid standards, Observer Variation, Proteomics organization & administration, Proteomics standards, Reference Standards, Reproducibility of Results, Research standards, Biomedical Research methods, Chromatography, Liquid methods, Proteomics methods
- Abstract
Despite the maturity reached by targeted proteomic strategies, reliable and standardized protocols are urgently needed to enhance reproducibility among different laboratories and analytical platforms, facilitating a more widespread use in biomedical research. To achieve this goal, the use of dimensionless relative retention times (iRT), defined on the basis of peptide standard retention times (RT), has lately emerged as a powerful tool. The robustness, reproducibility and utility of this strategy were examined for the first time in a multicentric setting, involving 28 laboratories that included 24 of the Spanish network of proteomics laboratories (ProteoRed-ISCIII). According to the results obtained in this study, dimensionless retention time values (iRTs) demonstrated to be a useful tool for transferring and sharing peptide retention times across different chromatographic set-ups both intra- and inter-laboratories. iRT values also showed very low variability over long time periods. Furthermore, parallel quantitative analyses showed a high reproducibility despite the variety of experimental strategies used, either MRM (multiple reaction monitoring) or pseudoMRM, and the diversity of analytical platforms employed., Biological Significance: From the very beginning of proteomics as an analytical science there has been a growing interest in developing standardized methods and experimental procedures in order to ensure the highest quality and reproducibility of the results. In this regard, the recent (2012) introduction of the dimensionless retention time concept has been a significant advance. In our multicentric (28 laboratories) study we explore the usefulness of this concept in the context of a targeted proteomics experiment, demonstrating that dimensionless retention time values is a useful tool for transferring and sharing peptide retention times across different chromatographic set-ups., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
35. Drafting the proteome landscape of myeloid-derived suppressor cells.
- Author
-
Gato M, Blanco-Luquin I, Zudaire M, de Morentin XM, Perez-Valderrama E, Zabaleta A, Kochan G, Escors D, Fernandez-Irigoyen J, and Santamaría E
- Subjects
- Animals, Humans, Immunotherapy, Neoplasms immunology, Neoplasms metabolism, Neoplasms therapy, Proteomics, Tumor Microenvironment, Myeloid Cells metabolism, Proteome metabolism
- Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that are defined by their myeloid origin, immature state, and ability to potently suppress T-cell responses. They regulate immune responses and the population significantly increases in the tumor microenvironment of patients with glioma and other malignant tumors. For their study, MDSCs are usually isolated from the spleen or directly of tumors from a large number of tumor-bearing mice although promising ex vivo differentiated MDSC production systems have been recently developed. During the last years, proteomics has emerged as a powerful approach to analyze MDSCs proteomes using shotgun-based mass spectrometry (MS), providing functional information about cellular homeostasis and metabolic state at a global level. Here, we will revise recent proteome profiling studies performed in MDSCs from different origins. Moreover, we will perform an integrative functional analysis of the protein compilation derived from these large-scale proteomic studies in order to obtain a comprehensive view of MDSCs biology. Finally, we will also discuss the potential application of high-throughput proteomic approaches to study global proteome dynamics and post-translational modifications (PTMs) during the differentiation process of MDSCs that will greatly boost the identification of novel MDSC-specific therapeutic targets to apply in cancer immunotherapy., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
36. Olfactory bulb proteome dynamics during the progression of sporadic Alzheimer's disease: identification of common and distinct olfactory targets across Alzheimer-related co-pathologies.
- Author
-
Zelaya MV, Pérez-Valderrama E, de Morentin XM, Tuñon T, Ferrer I, Luquin MR, Fernandez-Irigoyen J, and Santamaría E
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease pathology, Autopsy, Blotting, Western, Brain metabolism, Brain pathology, Dementia metabolism, Dementia pathology, Disease Progression, Female, Humans, Immunohistochemistry, Male, Mass Spectrometry, Middle Aged, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Protein Interaction Maps, Supranuclear Palsy, Progressive metabolism, Supranuclear Palsy, Progressive pathology, Alzheimer Disease metabolism, Olfactory Bulb metabolism, Proteome metabolism, Proteomics methods
- Abstract
Olfactory dysfunction is present in up to 90% of Alzheimer's disease (AD) patients. Although deposition of hyperphosphorylated tau and β-amyloid substrates are present in olfactory areas, the molecular mechanisms associated with decreased smell function are not completely understood. We have applied mass spectrometry-based quantitative proteomics to probe additional molecular disturbances in postmortem olfactory bulbs (OB) dissected from AD cases respect to neurologically intact controls (n=20, mean age 82.1 years). Relative proteome abundance measurements have revealed protein interaction networks progressively disturbed across AD stages suggesting an early imbalance in splicing factors, subsequent interrupted cycling of neurotransmitters, alteration in toxic and protective mechanisms of β-amyloid, and finally, a mitochondrial dysfunction together with disturbance in neuron-neuron adhesion. We also present novel molecular findings in the OB in an autopsy cohort composed by Lewy body disease (LBD), frontotemporal lobar degeneration (FTLD), mixed dementia, and progressive supranuclear palsy (PSP) cases (n = 41, mean age 79.7 years). Olfactory mediators deregulated during the progression of AD such as Visinin-like protein 1, RUFY3 protein, and Copine 6 were also differentially modulated in the OB in LBD, FTLD, and mixed dementia. Only Dipeptidyl aminopeptidase-like protein 6 showed a specific down-regulation in AD. However, no differences were observed in the olfactory expression of this protein panel in PSP subjects. This study demonstrates an olfactory progressive proteome modulation in AD, unveiling cross-disease similarities and differences especially for specific proteins involved in dendritic and axonic distributions that occur in the OB during the neurodegenerative process.
- Published
- 2015
- Full Text
- View/download PDF
37. Toward defining the anatomo-proteomic puzzle of the human brain: An integrative analysis.
- Author
-
Fernandez-Irigoyen J, Labarga A, Zabaleta A, de Morentin XM, Perez-Valderrama E, Zelaya MV, and Santamaria E
- Subjects
- Cerebrospinal Fluid metabolism, Humans, Mass Spectrometry, Brain metabolism, Computational Biology methods, Proteomics methods
- Abstract
The human brain is exceedingly complex, constituted by billions of neurons and trillions of synaptic connections that, in turn, define ∼900 neuroanatomical subdivisions in the adult brain (Hawrylycz et al. An anatomically comprehensive atlas of the human brain transcriptome. Nature 2012, 489, 391-399). The human brain transcriptome has revealed specific regional transcriptional signatures that are regulated in a spatiotemporal manner, increasing the complexity of the structural and molecular organization of this organ (Kang et al. Spatio-temporal transcriptome of the human brain. Nature 2011, 478, 483-489). During the last decade, neuroproteomics has emerged as a powerful approach to profile neural proteomes using shotgun-based MS, providing complementary information about protein content and function at a global level. Here, we revise recent proteome profiling studies performed in human brain, with special emphasis on proteome mapping of anatomical macrostructures, specific subcellular compartments, and cerebrospinal fluid. Moreover, we have performed an integrative functional analysis of the protein compilation derived from these large-scale human brain proteomic studies in order to obtain a comprehensive view of human brain biology. Finally, we also discuss the potential contribution of our meta-analysis to the Chromosome-centric Human Proteome Project initiative., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
38. A core of kinase-regulated interactomes defines the neoplastic MDSC lineage.
- Author
-
Gato-Cañas M, Martinez de Morentin X, Blanco-Luquin I, Fernandez-Irigoyen J, Zudaire I, Liechtenstein T, Arasanz H, Lozano T, Casares N, Chaikuad A, Knapp S, Guerrero-Setas D, Escors D, Kochan G, and Santamaría E
- Subjects
- Animals, Bone Marrow Cells cytology, Cell Differentiation, Cell Lineage, Cell Survival, Dendritic Cells cytology, Electric Impedance, Extracellular Signal-Regulated MAP Kinases metabolism, Gene Expression Profiling, HEK293 Cells, Humans, Inhibitory Concentration 50, Melanoma, Experimental, Mice, Mice, Inbred C57BL, Myeloid Cells immunology, Neoplasms metabolism, Phosphatidylinositol 3-Kinases metabolism, Proteasome Endopeptidase Complex metabolism, Proteomics, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering metabolism, Signal Transduction, Gene Expression Regulation, Neoplastic, Myeloid Cells cytology
- Abstract
Myeloid-derived suppressor cells (MDSCs) differentiate from bone marrow precursors, expand in cancer-bearing hosts and accelerate tumor progression. MDSCs have become attractive therapeutic targets, as their elimination strongly enhances anti-neoplastic treatments. Here, immature myeloid dendritic cells (DCs), MDSCs modeling tumor-infiltrating subsets or modeling non-cancerous (NC)-MDSCs were compared by in-depth quantitative proteomics. We found that neoplastic MDSCs differentially expressed a core of kinases which controlled lineage-specific (PI3K-AKT and SRC kinases) and cancer-induced (ERK and PKC kinases) protein interaction networks (interactomes). These kinases contributed to some extent to myeloid differentiation. However, only AKT and ERK specifically drove MDSC differentiation from myeloid precursors. Interfering with AKT and ERK with selective small molecule inhibitors or shRNAs selectively hampered MDSC differentiation and viability. Thus, we provide compelling evidence that MDSCs constitute a distinct myeloid lineage distinguished by a "kinase signature" and well-defined interactomes. Our results define new opportunities for the development of anti-cancer treatments targeting these tumor-promoting immune cells.
- Published
- 2015
- Full Text
- View/download PDF
39. A highly efficient tumor-infiltrating MDSC differentiation system for discovery of anti-neoplastic targets, which circumvents the need for tumor establishment in mice.
- Author
-
Liechtenstein T, Perez-Janices N, Gato M, Caliendo F, Kochan G, Blanco-Luquin I, Van der Jeught K, Arce F, Guerrero-Setas D, Fernandez-Irigoyen J, Santamaria E, Breckpot K, and Escors D
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Computational Biology methods, Dendritic Cells cytology, Flow Cytometry, Humans, Immunoblotting, Mice, Mice, Inbred C57BL, Cell Culture Techniques methods, Melanoma immunology, Myeloid Cells cytology, Proteomics methods
- Abstract
Myeloid-derived suppressor cells (MDSCs) exhibit potent immunosuppressive activities in cancer. MDSCs infiltrate tumors and strongly inhibit cancer-specific cytotoxic T cells. Their mechanism of differentiation and identification of MDSC-specific therapeutic targets are major areas of interest. We have devised a highly efficient and rapid method to produce very large numbers of melanoma-infiltrating MDSCs ex vivo without inducing tumors in mice. These MDSCs were used to study their differentiation, immunosuppressive activities and were compared to non-neoplastic counterparts and conventional dendritic cells using unbiased systems biology approaches. Differentially activated/deactivated pathways caused by cell type differences and by the melanoma tumor environment were identified. MDSCs increased the expression of trafficking receptors to sites of inflammation, endocytosis, changed lipid metabolism, and up-regulated detoxification pathways such as the expression of P450 reductase. These studies uncovered more than 60 potential novel therapeutic targets. As a proof of principle, we demonstrate that P450 reductase is the target of pro-drugs such as Paclitaxel, which depletes MDSCs following chemotherapy in animal models of melanoma and in human patients. Conversely, P450 reductase protects MDSCs against the cytotoxic actions of other chemotherapy drugs such as Irinotecan, which is ineffective for the treatment of melanoma.
- Published
- 2014
- Full Text
- View/download PDF
40. Molecular profiling of hepatocellular carcinoma in mice with a chronic deficiency of hepatic s-adenosylmethionine: relevance in human liver diseases.
- Author
-
Santamaría E, Muñoz J, Fernandez-Irigoyen J, Sesma L, Mora MI, Berasain C, Lu SC, Mato JM, Prieto J, Avila MA, and Corrales FJ
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Carcinoma, Hepatocellular pathology, Case-Control Studies, Down-Regulation, Electrophoresis, Gel, Two-Dimensional, Gene Expression Regulation, Neoplastic, Genetic Variation, Humans, Liver Diseases pathology, Liver Neoplasms pathology, Mass Spectrometry, Mice, Mice, Knockout, Middle Aged, Peptide Mapping, Protein Array Analysis, S-Adenosylmethionine genetics, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Trypsin pharmacology, Tumor Burden, Up-Regulation, Carcinoma, Hepatocellular metabolism, Liver Diseases metabolism, Liver Neoplasms metabolism, Proteome analysis, S-Adenosylmethionine deficiency
- Abstract
S-adenosylmethionine arises as a central molecule in the preservation of liver homeostasis as a chronic hepatic deficiency results in spontaneous development of steatohepatitis and hepatocellular carcinoma. In the present work, we have attempted a comprehensive analysis of proteins associated with hepatocarcinogenesis in MAT1A knock out mice using a combination of two-dimensional electrophoresis and mass spectrometry, to then apply the resulting information to identify hallmarks of human HCC. Our results suggest the existence of individual-specific factors that might condition the development of preneoplastic lesions. Proteomic analysis allowed the identification of 151 differential proteins in MAT1A-/- mice tumors. Among all differential proteins, 27 changed in at least 50% of the analyzed tumors, and some of these alterations were already detected months before the development of HCC in the KO liver. The expression level of genes coding for 13 of these proteins was markedly decreased in human HCC. Interestingly, seven of these genes were also found to be down-regulated in a pretumoral condition such as cirrhosis, while depletion of only one marker was assessed in less severe liver disorders.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.