85 results on '"Santofimia-Castaño P"'
Search Results
2. Targeting NUPR1-dependent stress granules formation to induce synthetic lethality in KrasG12D-driven tumors
- Author
-
Santofimia-Castaño, Patricia, Fraunhoffer, Nicolas, Liu, Xi, Bessone, Ivan Fernandez, di Magliano, Marina Pasca, Audebert, Stephane, Camoin, Luc, Estaras, Matias, Brenière, Manon, Modesti, Mauro, Lomberk, Gwen, Urrutia, Raul, Soubeyran, Philippe, Neira, Jose Luis, and Iovanna, Juan
- Published
- 2024
- Full Text
- View/download PDF
3. Stroma-targeting strategies in pancreatic cancer: a double-edged sword
- Author
-
Liu, Xi, Iovanna, Juan, and Santofimia-Castaño, Patricia
- Published
- 2023
- Full Text
- View/download PDF
4. Melatonin controls cell proliferation and modulates mitochondrial physiology in pancreatic stellate cells
- Author
-
Estaras, Matias, Ortiz-Placin, Candido, Castillejo-Rufo, Alba, Fernandez-Bermejo, Miguel, Blanco, Gerardo, Mateos, Jose M., Vara, Daniel, Gonzalez-Cordero, Pedro L., Chamizo, Sandra, Lopez, Diego, Rojas, Adela, Jaen, Isabel, de Armas, Noelia, Salido, Gines M., Iovanna, Juan L., Santofimia-Castaño, Patricia, and Gonzalez, Antonio
- Published
- 2023
- Full Text
- View/download PDF
5. NUPR1 protects against hyperPARylation-dependent cell death
- Author
-
Santofimia-Castaño, Patricia, Huang, Can, Liu, Xi, Xia, Yi, Audebert, Stephane, Camoin, Luc, Peng, Ling, Lomberk, Gwen, Urrutia, Raul, Soubeyran, Philippe, Neira, Jose Luis, and Iovanna, Juan
- Published
- 2022
- Full Text
- View/download PDF
6. Targeting intrinsically disordered proteins involved in cancer
- Author
-
Santofimia-Castaño, Patricia, Rizzuti, Bruno, Xia, Yi, Abian, Olga, Peng, Ling, Velázquez-Campoy, Adrián, Neira, José L., and Iovanna, Juan
- Published
- 2020
- Full Text
- View/download PDF
7. NUPR1 inhibitor ZZW-115 induces ferroptosis in a mitochondria-dependent manner
- Author
-
Huang, Can, Santofimia-Castaño, Patricia, Liu, Xi, Xia, Yi, Peng, Ling, Gotorbe, Célia, Neira, Jose Luis, Tang, Daolin, Pouyssegur, Jacques, and Iovanna, Juan
- Published
- 2021
- Full Text
- View/download PDF
8. Melatonin induces reactive oxygen species generation and changes in glutathione levels and reduces viability in human pancreatic stellate cells
- Author
-
Estaras, Matias, Moreno, Noelia, Santofimia-Castaño, Patricia, Martinez-Morcillo, Salome, Roncero, Vicente, Blanco, Gerardo, Lopez, Diego, Fernandez-Bermejo, Miguel, Mateos, Jose M., Iovanna, Juan L., Salido, Gines M., and Gonzalez, Antonio
- Published
- 2019
- Full Text
- View/download PDF
9. Melatonin modulates red-ox state and decreases viability of rat pancreatic stellate cells
- Author
-
Gonzalez, Antonio, Estaras, Matias, Martinez-Morcillo, Salome, Martinez, Remigio, García, Alfredo, Estévez, Mario, Santofimia-Castaño, Patricia, Tapia, Jose A., Moreno, Noelia, Pérez-López, Marcos, Míguez, María P., Blanco-Fernández, Gerardo, Lopez-Guerra, Diego, Fernandez-Bermejo, Miguel, Mateos, Jose M., Vara, Daniel, Roncero, Vicente, and Salido, Gines M.
- Published
- 2020
- Full Text
- View/download PDF
10. Inactivation of NUPR1 promotes cell death by coupling ER-stress responses with necrosis
- Author
-
Santofimia-Castaño, Patricia, Lan, Wenjun, Bintz, Jennifer, Gayet, Odile, Carrier, Alice, Lomberk, Gwen, Neira, José Luis, González, Antonio, Urrutia, Raul, Soubeyran, Philippe, and Iovanna, Juan
- Published
- 2018
- Full Text
- View/download PDF
11. E2F signature is predictive for the pancreatic adenocarcinoma clinical outcome and sensitivity to E2F inhibitors, but not for the response to cytotoxic-based treatments
- Author
-
Lan, Wenjun, Bian, Benjamin, Xia, Yi, Dou, Samir, Gayet, Odile, Bigonnet, Martin, Santofimia-Castaño, Patricia, Cong, Mei, Peng, Ling, Dusetti, Nelson, and Iovanna, Juan
- Published
- 2018
- Full Text
- View/download PDF
12. Interferences of resveratrol with fura-2-derived fluorescence in intracellular free-Ca2+ concentration determinations
- Author
-
Santofimia-Castaño, Patricia, Salido, Gines M., and Gonzalez, Antonio
- Published
- 2016
- Full Text
- View/download PDF
13. Melatonin induces calcium mobilization and influences cell proliferation independently of MT1/MT2 receptor activation in rat pancreatic stellate cells
- Author
-
Santofimia-Castaño, Patricia, Garcia-Sanchez, Lourdes, Ruy, Deborah Clea, Sanchez-Correa, Beatriz, Fernandez-Bermejo, Miguel, Tarazona, Raquel, Salido, Gines M., and Gonzalez, Antonio
- Published
- 2015
- Full Text
- View/download PDF
14. Intrinsically Disordered Chromatin Protein NUPR1 Binds to the Enzyme PADI4
- Author
-
Araujo-Abad, S., Neira, J. L., Rizzuti, B., García-Morales, P., Juan Romero, C. de, Santofimia-Castaño, P., and Iovanna, J.
- Subjects
Structural Biology ,Molecular Biology - Abstract
The nuclear protein 1 (NUPR1) is an intrinsically disordered protein involved in stress-mediated cellular conditions. Its paralogue nuclear protein 1-like (NUPR1L) is p53-regulated, and its expression down-regulates that of the NUPR1 gene. Peptidyl-arginine deiminase 4 (PADI4) is an isoform of a family of enzymes catalyzing arginine to citrulline conversion; it is also involved in stress-mediated cellular conditions. We characterized the interaction between NUPR1 and PADI4 in vitro, in silico, and in cellulo. The interaction of NUPR1 and PADI4 occurred with a dissociation constant of 18 ± 6 μM. The binding region of NUPR1, mapped by NMR, was a hydrophobic polypeptide patch surrounding the key residue Ala33, as pinpointed by: (i) computational results; and, (ii) site-directed mutagenesis of residues of NUPR1. The association between PADI4 and wild-type NUPR1 was also assessed in cellulo by using proximity ligation assays (PLAs) and immunofluorescence (IF), and it occurred mainly in the nucleus. Moreover, binding between NUPR1L and PADI4 also occurred in vitro with an affinity similar to that of NUPR1. Molecular modelling provided information on the binding hot spot for PADI4. This is an example of a disordered partner of PADI4, whereas its other known interacting proteins are well-folded. Altogether, our results suggest that the NUPR1/PADI4 complex could have crucial functions in modulating DNA-repair, favoring metastasis, or facilitating citrullination of other proteins.
- Published
- 2023
- Full Text
- View/download PDF
15. Pharmacological dose of melatonin reduces cytosolic calcium load in response to cholecystokinin in mouse pancreatic acinar cells
- Author
-
Santofimia-Castaño, Patricia, Ruy, Deborah Clea, Fernandez-Bermejo, Miguel, Salido, Gines M., and Gonzalez, Antonio
- Published
- 2014
- Full Text
- View/download PDF
16. Astrocytic NMDA receptors sustains antioxidant protection of neurons through a novel Cdk5-Nrf2 pathway: S18-01
- Author
-
Jimenez-Blasco, D., Santofimia-Castaño, P., Gonzalez, A., Almeida, A., and Bolanos, J. P.
- Published
- 2015
17. Melatonin modulates Ca2+ mobilization and amylase release in response to cholecystokinin octapeptide in mouse pancreatic acinar cells
- Author
-
Santofimia-Castaño, Patricia, Ruy, Deborah Clea, Salido, Ginés M., and González, Antonio
- Published
- 2013
- Full Text
- View/download PDF
18. Cinnamtannin B-1, a natural antioxidant that reduces the effects of H2O2 on CCK-8-evoked responses in mouse pancreatic acinar cells
- Author
-
Gonzalez, Antonio, Santofimia-Castaño, Patricia, Rivera-Barreno, Ramon, and Salido, Gines M.
- Published
- 2012
- Full Text
- View/download PDF
19. Resveratrol mobilizes Ca2+ from intracellular stores and induces c-Jun N-terminal kinase activation in tumoral AR42J cells
- Author
-
Garcia-Sanchez, Lourdes, Santofimia-Castaño, Patricia, Miro-Moran, Alvaro, Tapia, Jose A., Salido, Gines M., and Gonzalez, Antonio
- Published
- 2012
- Full Text
- View/download PDF
20. Iron-Sensitive Prodrugs That Trigger Active Ferroptosis in Drug-Tolerant Pancreatic Cancer Cells.
- Author
-
Antoszczak, Michał, Müller, Sebastian, Cañeque, Tatiana, Colombeau, Ludovic, Dusetti, Nelson, Santofimia-Castaño, Patricia, Gaillet, Christine, Puisieux, Alain, Iovanna, Juan Lucio, and Rodriguez, Raphaël
- Published
- 2022
- Full Text
- View/download PDF
21. Targeting NUPR1 by ZZW-115, alone and in combination with genotoxic agents, is an efficient strategy for PDAC treatment
- Author
-
Santofimia-Castaño, P., primary, Lan, W., additional, Swayden, M., additional, Xia, Y., additional, Zhou, Z., additional, Audebert, S., additional, Camoin, L., additional, Huang, C., additional, Peng, L., additional, Geli, V., additional, Velazquez-Campoy, A., additional, Abian, O., additional, Lomberk, G., additional, Urrutia, R., additional, Rizzuti, B., additional, Soubeyran, P., additional, Neira, J., additional, and Iovanna, J., additional
- Published
- 2020
- Full Text
- View/download PDF
22. Inducing ferroptosis by the NUPR1 inhibitor ZZW115 to kill pancreatic cancer cells
- Author
-
Huang, C., primary, Santofimia-Castaño, P., additional, Lan, W., additional, Fraunhoffer, N., additional, Meilerman, A., additional, and Iovanna, J., additional
- Published
- 2020
- Full Text
- View/download PDF
23. ZZW-115-dependent inhibition of NUPR1 nuclear translocation sensitizes cancer cells to genotoxic agents
- Author
-
Ligue Nationale contre le Cancer (France), Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), Diputación General de Aragón, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (España), National Natural Science Foundation of China, Agence Nationale de la Recherche (France), Lan, W., Santofimia-Castaño, P., Swayden, M., Xia, Y., Zhou, Z., Audebert, S., Camoin, L., Huang, C., Peng, L., Jiménez-Alesanco, Ana, Velázquez-Campoy, Adrián, Abian, Olga, Lomberk, Gwen, Urrutia, R., Rizzuti, Bruno, Geli, V., Soubeyran, P., Neira, José L., Iovanna, Juan Lucio, Ligue Nationale contre le Cancer (France), Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), Diputación General de Aragón, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (España), National Natural Science Foundation of China, Agence Nationale de la Recherche (France), Lan, W., Santofimia-Castaño, P., Swayden, M., Xia, Y., Zhou, Z., Audebert, S., Camoin, L., Huang, C., Peng, L., Jiménez-Alesanco, Ana, Velázquez-Campoy, Adrián, Abian, Olga, Lomberk, Gwen, Urrutia, R., Rizzuti, Bruno, Geli, V., Soubeyran, P., Neira, José L., and Iovanna, Juan Lucio
- Abstract
Establishing the interactome of the cancer-associated stress protein Nuclear Protein 1 (NUPR1), we found that it binds to several hundreds of proteins, including proteins involved in nuclear translocation, DNA repair, and key factors of the SUMO pathway. We demonstrated that the NUPR1 inhibitor ZZW-115, an organic synthetic molecule, competes with importins for the binding to the NLS region of NUPR1, thereby inhibiting its nuclear translocation. We hypothesized, and then proved, that inhibition of NUPR1 by ZZW-115 sensitizes cancer cells to DNA damage induced by several genotoxic agents. Strikingly, we found that treatment with ZZW-115 reduced SUMOylation of several proteins involved in DNA damage response (DDR). We further report that the presence of recombinant NUPR1 improved the SUMOylation in a cell-free system, indicating that NUPR1 directly stimulates the SUMOylation machinery. We propose that ZZW-115 sensitizes cancer cells to genotoxic agents by inhibiting the nuclear translocation of NUPR1 and thereby decreasing the SUMOylation-dependent functions of key proteins involved in the DDR.
- Published
- 2020
24. Targeting intrinsically disordered proteins involved in cancer
- Author
-
Canceropole PACA, Institut National de la Santé et de la Recherche Médicale (France), Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Gobierno de Aragón, National Natural Science Foundation of China, Fondation de France, Ministerio de Ciencia, Innovación y Universidades (España), SCOAP, Santofimia-Castaño, P., Rizzuti, Bruno, Xia, Y., Abian, Olga, Peng, L., Velázquez-Campoy, Adrián, Neira, José L., Iovanna, Juan Lucio, Canceropole PACA, Institut National de la Santé et de la Recherche Médicale (France), Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Gobierno de Aragón, National Natural Science Foundation of China, Fondation de France, Ministerio de Ciencia, Innovación y Universidades (España), SCOAP, Santofimia-Castaño, P., Rizzuti, Bruno, Xia, Y., Abian, Olga, Peng, L., Velázquez-Campoy, Adrián, Neira, José L., and Iovanna, Juan Lucio
- Abstract
Intrinsically disordered proteins (IDPs) do not have a well-defined structure under physiological conditions, but they have key roles in cell signaling and regulation, and they are frequently related to the development of diseases, such as cancer and other malignancies. This has converted IDPs in attractive therapeutic targets; however, targeting IDPs is challenging because of their dynamic nature. In the last years, different experimental and computational approaches, as well as the combination of both, have been explored to identify molecules to target either the hot-spots or the allosteric sites of IDPs. In this review, we summarize recent developments in successful targeting of IDPs, all of which are involved in different cancer types. The strategies used to develop and design (or in one particular example, to repurpose) small molecules targeting IDPs are, in a global sense, similar to those used in well-folded proteins: (1) screening of chemically diverse or target-oriented compound libraries; or (2) study of the interfaces involved in recognition of their natural partners, and design of molecular candidates capable of binding to such binding interface. We describe the outcomes of using these approaches in targeting IDPs involved in cancer, in the view to providing insight, to target IDPs in general. In a broad sense, the designed small molecules seem to target the most hydrophobic regions of the IDPs, hampering macromolecule (DNA or protein)–IDP interactions; furthermore, in most of the molecule–IDP complexes described so far, the protein remains disordered.
- Published
- 2019
25. Dendrimers as Competitors of Protein–Protein Interactions of the Intrinsically Disordered Nuclear Chromatin Protein NUPR1.
- Author
-
Neira, José L., Correa, Juan, Rizzuti, Bruno, Santofimia-Castaño, Patricia, Abian, Olga, Velázquez-Campoy, Adrián, Fernandez-Megia, Eduardo, and Iovanna, Juan L.
- Published
- 2019
- Full Text
- View/download PDF
26. Astrocyte NMDA receptors' activity sustains neuronal survival through a Cdk5–Nrf2 pathway
- Author
-
Jimenez-Blasco, D, primary, Santofimia-Castaño, P, additional, Gonzalez, A, additional, Almeida, A, additional, and Bolaños, J P, additional
- Published
- 2015
- Full Text
- View/download PDF
27. Modulation of copper accumulation and copper-induced toxicity by antioxidants and copper chelators in cultured primary brain astrocytes.
- Author
-
Bulcke, Felix, Santofimia-Castaño, Patricia, Gonzalez-Mateos, Antonio, and Dringen, Ralf
- Subjects
COPPER bioaccumulation ,COPPER poisoning ,CELL survival ,PHYSIOLOGICAL effects of antioxidants ,CELL culture ,ASTROCYTES ,BRAIN physiology - Abstract
Copper is essential for several important cellular processes, but an excess of copper can also lead to oxidative damage. In brain, astrocytes are considered to play a pivotal role in the copper homeostasis and antioxidative defence. To investigate whether antioxidants and copper chelators can modulate the uptake and the toxicity of copper ions in brain astrocytes, we used primary astrocytes as cell culture model. These cells accumulated substantial amounts of copper during exposure to copper chloride. Copper accumulation was accompanied by a time- and concentration-dependent loss in cell viability, as demonstrated by a lowering in cellular MTT reduction capacity and by an increase in membrane permeability for propidium iodide. During incubations in the presence of the antioxidants ascorbate, trolox or ebselen, the specific cellular copper content and the toxicity in copper chloride-treated astrocyte cultures were strongly increased. In contrast, the presence of the copper chelators bathocuproine disulfonate or tetrathiomolybdate lowered the cellular copper accumulation and the copper-induced as well as the ascorbate-accelerated copper toxicity was fully prevented. These data suggest that predominantly the cellular content of copper determines copper-induced toxicity in brain astrocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
28. The natural antioxidant cinnamtannin B-1 reduces the effects of H2O2 on CCK-8-evoked responses in mouse pancreatic acinar cells.
- Author
-
Salido, G. M., Santofimia-Castaño, P., Rivera-Barreno, R., and Gonzalez, A.
- Published
- 2011
29. Kainate-evoked glutamate secretion is reduced by ethanol in rat hippocampal astrocytes.
- Author
-
Santofimia-Castaño, P., Portilla-Cuenca, J. C., Salido, G. M., and Gonzalez, A.
- Published
- 2011
30. Melatonin modulates metabolic adaptation of pancreatic stellate cells subjected to hypoxia.
- Author
-
Estaras M, Martinez R, Garcia A, Ortiz-Placin C, Iovanna JL, Santofimia-Castaño P, and Gonzalez A
- Subjects
- Actins metabolism, Cells, Cultured, Collagen metabolism, Fibrosis, Humans, Hypoxia metabolism, Pancreas metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, TOR Serine-Threonine Kinases metabolism, Melatonin metabolism, Melatonin pharmacology, Pancreatic Stellate Cells metabolism, Pancreatic Stellate Cells pathology
- Abstract
Pancreatic stellate cells (PSCs), the main cell type responsible for the development of fibrosis in pancreatic cancer, proliferate actively under hypoxia. Melatonin has received attention as a potential antifibrotic agent due to its anti-proliferative actions on PSCs. In this work, we investigated the activation of the PI3K/Akt/mTOR pathway and the metabolic adaptations that PSCs undergo under hypoxic conditions, as well as the probable modulation by melatonin. Incubation of cells under hypoxia induced an increase in cell proliferation, and in the expression of alpha-smooth muscle actin and of collagen type 1. In addition, an increase in the phosphorylation of Akt was observed, whereas a decrease in the phosphorylation of PTEN and GSK-3b was noted. The phosphorylation of mTOR and its substrate p70 S6K was decreased under hypoxia. Treatment of PSCs with melatonin under hypoxia diminished cell proliferation, the levels of alpha-smooth muscle actin and of collagen type 1, the phosphorylation of Akt and increased phosphorylation of mTOR. Mitochondrial activity decreased in PSCs under hypoxia. A glycolytic shift was observed. Melatonin further decreased mitochondrial activity. Under hypoxia, no increase in autophagic flux was noted. However, melatonin treatment induced autophagy activation. Nevertheless, inhibition of this process did not induce detectable changes in the viability of cells treated with melatonin. We conclude that PSCs undergo metabolic adaptation under hypoxia that might help them survive and that pharmacological concentrations of melatonin modulate cell responses to hypoxia. Our results contribute to the knowledge of the mechanisms by which melatonin could modulate fibrosis within the pancreas., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. Design of Inhibitors of the Intrinsically Disordered Protein NUPR1: Balance between Drug Affinity and Target Function.
- Author
-
Rizzuti B, Lan W, Santofimia-Castaño P, Zhou Z, Velázquez-Campoy A, Abián O, Peng L, Neira JL, Xia Y, and Iovanna JL
- Subjects
- Adenocarcinoma pathology, Animals, Basic Helix-Loop-Helix Transcription Factors chemistry, Calorimetry, Humans, Intrinsically Disordered Proteins genetics, Ligands, Mice, Neoplasm Proteins chemistry, Piperazines chemical synthesis, Piperazines pharmacology, Thiazines chemical synthesis, Thiazines pharmacology, Trifluoperazine chemistry, Trifluoperazine pharmacology, Adenocarcinoma drug therapy, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Carcinoma, Pancreatic Ductal drug therapy, Intrinsically Disordered Proteins antagonists & inhibitors, Neoplasm Proteins antagonists & inhibitors, Piperazines chemistry, Thiazines chemistry
- Abstract
Intrinsically disordered proteins (IDPs) are emerging as attractive drug targets by virtue of their physiological ubiquity and their prevalence in various diseases, including cancer. NUPR1 is an IDP that localizes throughout the whole cell, and is involved in the development and progression of several tumors. We have previously repurposed trifluoperazine (TFP) as a drug targeting NUPR1 and, by using a ligand-based approach, designed the drug ZZW-115 starting from the TFP scaffold. Such derivative compound hinders the development of pancreatic ductal adenocarcinoma (PDAC) in mice, by hampering nuclear translocation of NUPR1. Aiming to further improve the activity of ZZW-115, here we have used an indirect drug design approach to modify its chemical features, by changing the substituent attached to the piperazine ring. As a result, we have synthesized a series of compounds based on the same chemical scaffold. Isothermal titration calorimetry (ITC) showed that, with the exception of the compound preserving the same chemical moiety at the end of the alkyl chain as ZZW-115, an increase of the length by a single methylene group (i.e., ethyl to propyl) significantly decreased the affinity towards NUPR1 measured in vitro, whereas maintaining the same length of the alkyl chain and adding heterocycles favored the binding affinity. However, small improvements of the compound affinity towards NUPR1, as measured by ITC, did not result in a corresponding improvement in their inhibitory properties and in cellulo functions, as proved by measuring three different biological effects: hindrance of the nuclear translocation of the protein, sensitization of cells against DNA damage mediated by NUPR1, and prevention of cancer cell growth. Our findings suggest that a delicate compromise between favoring ligand affinity and controlling protein function may be required to successfully design drugs against NUPR1, and likely other IDPs.
- Published
- 2021
- Full Text
- View/download PDF
32. NUPR1: A Critical Regulator of the Antioxidant System.
- Author
-
Huang C, Santofimia-Castaño P, and Iovanna J
- Abstract
Nuclear protein 1 (NUPR1) is a small intrinsically disordered protein (IDP) activated in response to various types of cellular stress, including endoplasmic reticulum (ER) stress and oxidative stress. Reactive oxygen species (ROS) are mainly produced during mitochondrial oxidative metabolism, and directly impact redox homeostasis and oxidative stress. Ferroptosis is a ROS-dependent programmed cell death driven by an iron-mediated redox reaction. Substantial evidence supports a maintenance role of the stress-inducible protein NUPR1 on cancer cell metabolism that confers chemotherapeutic resistance by upregulating mitochondrial function-associated genes and various antioxidant genes in cancer cells. NUPR1, identified as an antagonist of ferroptosis, plays an important role in redox reactions. This review summarizes the current knowledge on the mechanism behind the observed impact of NUPR1 on mitochondrial function, energy metabolism, iron metabolism, and the antioxidant system. The therapeutic potential of genetic or pharmacological inhibition of NUPR1 in cancer is also discussed. Understanding the role of NUPR1 in the antioxidant system and the mechanisms behind its regulation of ferroptosis may promote the development of more efficacious strategies for cancer therapy.
- Published
- 2021
- Full Text
- View/download PDF
33. NUPR1 interacts with eIF2α and is required for resolution of the ER stress response in pancreatic tissue.
- Author
-
Borrello MT, Santofimia-Castaño P, Bocchio M, Listi A, Fraunhoffer N, Soubeyran P, Chevet E, Pin C, and Iovanna J
- Subjects
- Acinar Cells metabolism, Acinar Cells ultrastructure, Animals, Blotting, Western, Cell Line, Tumor, DNA-Binding Proteins metabolism, Eukaryotic Initiation Factor-2 metabolism, Humans, Mice, Knockout, Microscopy, Electron, Transmission, Neoplasm Proteins metabolism, Pancreas cytology, Pancreas ultrastructure, Protein Binding, Reverse Transcriptase Polymerase Chain Reaction, Mice, DNA-Binding Proteins genetics, Endoplasmic Reticulum Stress genetics, Eukaryotic Initiation Factor-2 genetics, Gene Expression Regulation, Neoplasm Proteins genetics, Pancreas metabolism, Unfolded Protein Response genetics
- Abstract
Nuclear protein 1 (NUPR1) is a stress response protein overexpressed upon cell injury in virtually all organs including the exocrine pancreas. Despite NUPR1's well-established role in the response to cell stress, the molecular and structural machineries triggered by NUPR1 activation remain largely debated. In this study, we uncover a new role for NUPR1, participating in the unfolded protein response (UPR) and the integrated stress response. Biochemical results and ultrastructural morphological observations revealed alterations in the UPR of acinar cells of germline-deleted NUPR1 murine models, consistent with the inability to restore general protein synthesis after stress induction. Bioinformatic analysis of NUPR1-interacting partners showed significant enrichment in translation initiation factors, including eukaryotic initiation factor (eIF) 2α. Co-immunoprecipitation and proximity ligation assays confirmed the interaction between NUPR1 and eIF2α and its phosphorylated form (p-eIF2α). Furthermore, our data suggest loss of NUPR1 in cells results in maintained eIF2α phosphorylation and evaluation of nascent proteins by click chemistry revealed that NUPR1-depleted PANC-1 cells displayed a slower poststress protein synthesis recovery when compared to wild-type. Combined, these data propose a novel role for NUPR1 in the integrated stress response pathway, at least partially through promoting efficient PERK branch activity and resolution through a unique interaction with eIF2α., (© 2021 Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
34. Targeting Fibrosis: The Bridge That Connects Pancreatitis and Pancreatic Cancer.
- Author
-
Huang C, Iovanna J, and Santofimia-Castaño P
- Subjects
- Animals, Cancer-Associated Fibroblasts pathology, Fibrosis, Humans, Molecular Targeted Therapy, Stress, Physiological, Pancreatic Neoplasms pathology, Pancreatitis pathology
- Abstract
Pancreatic fibrosis is caused by the excessive deposits of extracellular matrix (ECM) and collagen fibers during repeated necrosis to repair damaged pancreatic tissue. Pancreatic fibrosis is frequently present in chronic pancreatitis (CP) and pancreatic cancer (PC). Clinically, pancreatic fibrosis is a pathological feature of pancreatitis and pancreatic cancer. However, many new studies have found that pancreatic fibrosis is involved in the transformation from pancreatitis to pancreatic cancer. Thus, the role of fibrosis in the crosstalk between pancreatitis and pancreatic cancer is critical and still elusive; therefore, it deserves more attention. Here, we review the development of pancreatic fibrosis in inflammation and cancer, and we discuss the therapeutic strategies for alleviating pancreatic fibrosis. We further propose that cellular stress response might be a key driver that links fibrosis to cancer initiation and progression. Therefore, targeting stress proteins, such as nuclear protein 1 (NUPR1), could be an interesting strategy for pancreatic fibrosis and PC treatment.
- Published
- 2021
- Full Text
- View/download PDF
35. Combating pancreatic cancer chemoresistance by triggering multiple cell death pathways.
- Author
-
Santofimia-Castaño P and Iovanna J
- Subjects
- Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Humans, Treatment Failure, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Pancreatic Ductal drug therapy, Cell Death drug effects, Drug Resistance, Neoplasm drug effects, Pancreatic Neoplasms drug therapy
- Abstract
Pancreatic cancer is the fourth most common cause of cancer-associated death in western countries, where the incidence and number of deaths are increasing every year. Intrinsic or acquired resistance of tumor cells to chemotherapy agents is the major reason for failure of traditional cancer treatment. Several factors are implicated in this impressive resistance; however, of these, it is important to highlight the extensive cellular heterogeneity of these tumors. This heterogeneity is linked to a wide range of sensitivity that different clones in the same tumor display to chemotherapeutic agents. Accordingly, recent findings in this field have discovered new therapeutic targets in order to develop new combinatory treatments, as well as to induce several cell death pathways and reduce therapy-threshold and likelihood of future resistance. Accordingly, recent research has focused on targeting mitochondria, an organelle with key roles regulating cell death signaling pathways, such as apoptosis, necroptosis, autophagy, ferroptosis, or parthanatos. These findings - identifying new compounds, alone or in combination, that can target pancreatic ductal adenocarcinoma cell resistance - could be the key to future treatments., (Copyright © 2021 IAP and EPC. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
36. Response to the Letter to the editor regarding "Targeting NUPR1 with the small compound ZZW-115 is an efficient strategy to treat hepatocellular carcinoma" by Jiong Lin.
- Author
-
Santofimia-Castaño P, Lan W, Xia Y, Zhou Z, Huang C, Barea D, Cervello M, Giannitrapani L, Montalto G, Peng L, and Iovanna J
- Subjects
- Gene Expression Regulation, Neoplastic, Humans, Neoplasm Proteins metabolism, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular genetics, Liver Neoplasms drug therapy, Liver Neoplasms genetics
- Published
- 2021
- Full Text
- View/download PDF
37. Intrinsically disordered protein NUPR1 binds to the armadillo-repeat domain of Plakophilin 1.
- Author
-
Santofimia-Castaño P, Rizzuti B, Pey AL, Fárez-Vidal ME, Iovanna JL, and Neira JL
- Subjects
- Humans, Male, Carcinogenesis metabolism, Cell Adhesion physiology, Cell Line, Tumor, Desmosomes metabolism, Intrinsically Disordered Proteins metabolism, Magnetic Resonance Imaging methods, Molecular Docking Simulation methods, Protein Domains physiology, Tryptophan metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Neoplasm Proteins metabolism, Plakophilins metabolism, Protein Binding physiology
- Abstract
Plakophilin 1 (PKP1), a member of the armadillo repeat family of proteins, is a scaffold component of desmosomes, which are key structural components for cell-cell adhesion. However, PKP1 can be also found in the nucleus of several cells. NUPR1 is an intrinsically disordered protein (IDP) that localizes throughout the whole cell, and intervenes in the development and progression of several cancers. In this work, we studied the binding between PKP1 and NUPR1 by using several in vitro biophysical techniques and in cellulo approaches. The interaction occurred with an affinity in the low micromolar range (~10 μM), and involved the participation of at least one of the tryptophan residues of PKP1 (as shown by fluorescence and molecular docking). The binding region of NUPR1, mapped by NMR and molecular modelling, was a polypeptide patch at the 30s region of its sequence. The association between PKP1 and NUPR1 also occurred in cellulo and was localized in the nucleus, as tested by protein ligation assays (PLAs). We hypothesize that NUPR1 plays an active role in carcinogenesis modulating the function of PKP1., Competing Interests: Declaration of competing interest The authors declare no conflict of interest in regard to this manuscript., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
38. Evidencing a Pancreatic Ductal Adenocarcinoma Subpopulation Sensitive to the Proteasome Inhibitor Carfilzomib.
- Author
-
Fraunhoffer NA, Abuelafia AM, Bigonnet M, Gayet O, Roques J, Telle E, Santofimia-Castaño P, Borrello MT, Chuluyan E, Dusetti N, and Iovanna J
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma pathology, Adult, Aged, Apoptosis drug effects, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Lineage drug effects, Cell Lineage genetics, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Female, Humans, Male, Middle Aged, Molecular Targeted Therapy, Neoplasm Proteins genetics, Oligopeptides adverse effects, Prognosis, Proteasome Inhibitors adverse effects, Transcriptome drug effects, Unfolded Protein Response genetics, Adenocarcinoma drug therapy, Carcinoma, Pancreatic Ductal drug therapy, Oligopeptides pharmacology, Proteasome Inhibitors pharmacology, Transcriptome genetics
- Abstract
Purpose: Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a survival rate less than 5%. Multiple chemotherapeutic drugs have been tested to improve patient prognosis; however, the clinical efficacy of these treatments is low. One of the most controversial family of drugs are the proteasome inhibitors, which have displayed promising effects in preclinical studies, but low clinical performance. Here, we unravel a specific transcriptomic signature that discriminates a subgroup of patients sensitive to the proteasome inhibitor carfilzomib., Experimental Design: First, we identified a subpopulation of PDAC-derived primary cells cultures (PDPCC) sensitive to the proteasome inhibitor carfilzomib. Then, we selected a transcriptomic signature that predicts carfilzomib chemosensitivity using independent component analysis on the transcriptome of PDPCC. Finally, we validated the signature in an independent cohort of PDAC biopsy-derived pancreatic organoids., Results: Sensitive phenotype was characterized by a high expression of genes related with a cornified/squamous pathway and a downregulation of epithelial-mesenchymal transition genes. Interestingly, carfilzomib-sensitive transcriptomic profile did not show any association with the proteasome activity but strongly correlates with ATF4 and CHOP expression, which are key markers of the unfolded protein response and critical to trigger the cell death program. Concordantly, sensitive phenotype showed a high level of the de novo RNA and protein synthesis compared with the resistant one and, most important, cell death induced by carfilzomib is dependent of the translational activity., Conclusions: We demonstrate the existence of a carfilzomib-sensitive PDAC subgroup with a specific transcriptomic phenotype that could explain the biological reason for this responsiveness., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
39. ZZW-115-dependent inhibition of NUPR1 nuclear translocation sensitizes cancer cells to genotoxic agents.
- Author
-
Lan W, Santofimia-Castaño P, Swayden M, Xia Y, Zhou Z, Audebert S, Camoin L, Huang C, Peng L, Jiménez-Alesanco A, Velázquez-Campoy A, Abián O, Lomberk G, Urrutia R, Rizzuti B, Geli V, Soubeyran P, Neira JL, and Iovanna J
- Subjects
- Animals, Apoptosis, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Nucleus drug effects, Cell Proliferation, DNA Repair, Female, Humans, Mice, Mice, Inbred BALB C, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Protein Transport, Sumoylation, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Carcinoma, Pancreatic Ductal drug therapy, Cell Nucleus metabolism, DNA Damage, Gene Expression Regulation, Neoplastic drug effects, Neoplasm Proteins antagonists & inhibitors, Pancreatic Neoplasms drug therapy, Piperazines pharmacology, Thiazines pharmacology
- Abstract
Establishing the interactome of the cancer-associated stress protein Nuclear Protein 1 (NUPR1), we found that it binds to several hundreds of proteins, including proteins involved in nuclear translocation, DNA repair, and key factors of the SUMO pathway. We demonstrated that the NUPR1 inhibitor ZZW-115, an organic synthetic molecule, competes with importins for the binding to the NLS region of NUPR1, thereby inhibiting its nuclear translocation. We hypothesized, and then proved, that inhibition of NUPR1 by ZZW-115 sensitizes cancer cells to DNA damage induced by several genotoxic agents. Strikingly, we found that treatment with ZZW-115 reduced SUMOylation of several proteins involved in DNA damage response (DDR). We further report that the presence of recombinant NUPR1 improved the SUMOylation in a cell-free system, indicating that NUPR1 directly stimulates the SUMOylation machinery. We propose that ZZW-115 sensitizes cancer cells to genotoxic agents by inhibiting the nuclear translocation of NUPR1 and thereby decreasing the SUMOylation-dependent functions of key proteins involved in the DDR.
- Published
- 2020
- Full Text
- View/download PDF
40. Targeting NUPR1 with the small compound ZZW-115 is an efficient strategy to treat hepatocellular carcinoma.
- Author
-
Lan W, Santofimia-Castaño P, Xia Y, Zhou Z, Huang C, Fraunhoffer N, Barea D, Cervello M, Giannitrapani L, Montalto G, Peng L, and Iovanna J
- Subjects
- Adenosine Triphosphate metabolism, Adult, Aged, Amino Acid Chloromethyl Ketones pharmacology, Animals, Apoptosis drug effects, Basic Helix-Loop-Helix Transcription Factors analysis, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Female, Humans, Liver Neoplasms metabolism, Liver Neoplasms pathology, Male, Mice, Mice, Inbred BALB C, Middle Aged, Mitochondria drug effects, Mitochondria metabolism, Neoplasm Proteins analysis, Piperazines therapeutic use, Thiazines therapeutic use, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, Neoplasm Proteins antagonists & inhibitors, Piperazines pharmacology, Thiazines pharmacology
- Abstract
HCC is a highly lethal malignancy with Sorafenib as the only molecularly targeted drug. The multifunctional stress-associated protein, NUPR1, plays an essential role in controlling cell growth, migration, invasion and Sorafenib resistance in HCC. We report here that NUPR1 expression is absent in healthy liver and it is progressively upregulated in HCC premalignant lesions such as hepatitis and cirrhosis with a maximum expression in HCC samples, highlighting that NUPR1 is a potential drug target for HCC. We therefore assessed in this work, ZZW-115, a strong inhibitor of NUPR1, as a promising candidate for the treatment of HCC. We validated its extraordinary antitumor effect on HCC by using two HCC cell lines, HepG2-and Hep3B, both in cell based experiments and xenografted mice. We further revealed that ZZW-115 treatment induced cell death by apoptosis and necroptosis mechanisms, with a concomitant mitochondrial metabolism failure that triggers lower ATP production. Furthermore, the ATP depletion cannot be rescued by the apoptosis inhibitor Z-VAD-FMK and/or the necrosis inhibitor Necrostatin-1, indicating that ZZW-115 induces cell death through the mitochondrial failure., Competing Interests: Declaration of competing interest Authors declare not conflicts of interest., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
41. Dissecting the Anticancer Mechanism of Trifluoperazine on Pancreatic Ductal Adenocarcinoma.
- Author
-
Huang C, Lan W, Fraunhoffer N, Meilerman A, Iovanna J, and Santofimia-Castaño P
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers with almost no curative chemotherapeutic treatment. Besides the development of new compounds, repurposing of approved drugs to treat cancer, alone or in combination, has become an attractive strategy, showing many therapeutic and economic advantages. However, it is necessary to improve our knowledge about the mechanism of cell death elicited by approved drugs itself, but also to rationally develop more powerful multidrug treatments. In this work, we focus our attention on determining the mechanism promoting cell death following trifluoperazine (TFP) treatment, which is an antipsychotic drug with strong anticancer activity in PDAC. We demonstrate that TFP induces cell death by apoptosis and necroptosis, which can be partially inhibited by Z-VAD-FMK as well as necrostatin-1, respectively. This cell death promotion is triggered by a poor ATP content, observed in TFP-treated cells as a consequence of a dramatic decrease in OXPHOS metabolism due to mitochondrial stress. Remarkably, mitochondrial homeostasis was seriously affected, and a loss of mitochondrial membrane potential and ROS overproduction was observed. Moreover, this mitochondrial stress was coupled with an ER stress and the activation of the endoplasmic-reticulum-associated protein degradation (ERAD) and the unf olded protein response (UPR) pathways. We took advantage of this information and inhibited this process by using the proteasome inhibitors MG-132 or bortezomib compounds in combination with TFP and found a significant improvement of the anticancer effect of the TFP on primary PDAC-derived cells. In conclusion, this study not only uncovers the molecular mechanisms that are triggered upon TFP-treatment but also its possible combination with bortezomib for the future development of therapies for pancreatic cancer.
- Published
- 2019
- Full Text
- View/download PDF
42. Targeting the Stress-Induced Protein NUPR1 to Treat Pancreatic Adenocarcinoma.
- Author
-
Santofimia-Castaño P, Xia Y, Peng L, Velázquez-Campoy A, Abián O, Lan W, Lomberk G, Urrutia R, Rizzuti B, Soubeyran P, Neira JL, and Iovanna J
- Subjects
- Adenocarcinoma metabolism, Animals, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Dose-Response Relationship, Drug, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Neoplasm Proteins antagonists & inhibitors, Pancreatic Neoplasms metabolism, Phenothiazines pharmacology, Protein Transport drug effects, Xenograft Model Antitumor Assays, Adenocarcinoma drug therapy, Basic Helix-Loop-Helix Transcription Factors metabolism, Neoplasm Proteins metabolism, Pancreatic Neoplasms drug therapy, Phenothiazines therapeutic use
- Abstract
Cancer cells activate stress-response mechanisms to adapt themselves to a variety of stressful conditions. Among these protective mechanisms, those controlled by the stress-induced nuclear protein 1 (NUPR1 ) belong to the most conserved ones. NUPR1 is an 82-residue-long, monomeric, basic and intrinsically disordered protein (IDP), which was found to be invariably overexpressed in some, if not all, cancer tissues. Remarkably, we and others have previously showed that genetic inactivation of the Nupr1 gene antagonizes the growth of pancreatic cancer as well as several other tumors. With the use of a multidisciplinary strategy by combining biophysical, biochemical, bioinformatic, and biological approaches, a trifluoperazine-derived compound, named ZZW-115, has been identified as an inhibitor of the NUPR1 functions. The anticancer activity of the ZZW-115 was first validated on a large panel of cancer cells. Furthermore, ZZW-115 produced a dose-dependent tumor regression of the tumor size in xenografted mice. Mechanistically, we have demonstrated that NUPR1 binds to several importins. Because ZZW-115 binds NUPR1 through the region around the amino acid Thr68, which is located into the nuclear location signal (NLS) region of the protein, we demonstrated that treatment with ZZW-115 inhibits completely the translocation of NUPR1 from the cytoplasm to the nucleus by competing with importins., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
43. Designing and repurposing drugs to target intrinsically disordered proteins for cancer treatment: using NUPR1 as a paradigm.
- Author
-
Santofimia-Castaño P, Rizzuti B, Xia Y, Abian O, Peng L, Velázquez-Campoy A, Iovanna JL, and Neira JL
- Abstract
Intrinsically disordered proteins (IDPs) do not have a well-defined structure, but they have key biological tasks in cancer development. By using the disordered cancer-related protein NUPR1 as a proof-of-concept, we have developed a new multidisciplinary approach to tackle drug-design against IDPs, using it to repurpose drugs for treating pancreatic adenocarcinoma (PDAC).
- Published
- 2019
- Full Text
- View/download PDF
44. Ligand-based design identifies a potent NUPR1 inhibitor exerting anticancer activity via necroptosis.
- Author
-
Santofimia-Castaño P, Xia Y, Lan W, Zhou Z, Huang C, Peng L, Soubeyran P, Velázquez-Campoy A, Abián O, Rizzuti B, Neira JL, and Iovanna J
- Subjects
- Animals, Hep G2 Cells, Humans, Jurkat Cells, Mice, Mice, Nude, Neoplasms metabolism, Neoplasms pathology, PC-3 Cells, Xenograft Model Antitumor Assays, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Necroptosis drug effects, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Neoplasms drug therapy, Trifluoperazine analogs & derivatives, Trifluoperazine chemical synthesis, Trifluoperazine chemistry, Trifluoperazine pharmacology
- Abstract
Intrinsically disordered proteins (IDPs) are emerging as attractive drug targets by virtue of their prevalence in various diseases including cancer. Drug development targeting IDPs is challenging because they have dynamical structure features and conventional drug design is not applicable. NUPR1 is an IDP playing an important role in pancreatic cancer. We previously reported that Trifluoperazine (TFP), an antipsychotic agent, was capable of binding to NUPR1 and inhibiting tumors growth. Unfortunately, TFP showed strong central nervous system side-effects. In this work, we undertook a multidisciplinary approach to optimize TFP, based on the synergy of computer modeling, chemical synthesis, and a variety of biophysical, biochemical and biological evaluations. A family of TFP-derived compounds was produced and the most active one, named ZZW-115, showed a dose-dependent tumor regression with no neurological effects and induced cell death mainly by necroptosis. This study opens a new perspective for drug development against IDPs, demonstrating the possibility of successful ligand-based drug design for such challenging targets.
- Published
- 2019
- Full Text
- View/download PDF
45. α-lipoic acid reduces postreperfusion syndrome in human liver transplantation - a pilot study.
- Author
-
Casciato P, Ambrosi N, Caro F, Vazquez M, Müllen E, Gadano A, de Santibañes E, de Santibañes M, Zandomeni M, Chahdi M, Lazarte JC, Biagiola DA, Iaquinandi JC, Santofimia-Castaño P, Iovanna J, Incardona C, and Chuluyan E
- Subjects
- Aged, Alarmins metabolism, Apoptosis, Biopsy, Cold Ischemia, Cytokines metabolism, Double-Blind Method, Female, Follow-Up Studies, Humans, Hypoxia, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Inhibitor of Apoptosis Proteins metabolism, Male, Middle Aged, Oxidative Stress, Patient Safety, Pilot Projects, Reperfusion methods, Ubiquitin-Protein Ligases metabolism, Liver Transplantation, Reperfusion Injury prevention & control, Thioctic Acid pharmacology
- Abstract
A double-blind randomized controlled trial was performed to compare the safety and efficacy of α-lipoic acid (ALA) in liver transplantation (LT). The grafts were randomized to receive ALA or placebo before the cold ischemia time. Furthermore, patients transplanted with the ALA-perfused graft received 600 mg of intravenous ALA, while patients with the nonperfused graft received the placebo just before graft reperfusion. Hepatic biopsy was performed 2 h postreperfusion. Blood samples were collected before, during and 1 and 2 days after reperfusion. Quantitative polymerase chain reaction (qPCR) analysis was performed on biopsies to assess genes involved in the response to hypoxia, apoptosis, cell growth, survival and proliferation, cytokine production and tissue damage protection. Nine of 40 patients developed postreperfusion syndrome (PRS), but seven of them belonged to the control group. There was a decrease in PHD2 and an increase in alpha subunit of hypoxia-inducible factor-1 (HIF-1α) and baculoviral IAP repeat containing 2 (Birc2) transcript levels in the biopsies from the ALA-treated versus the control group of patients. Additionally, plasma levels of alarmins were lower in ALA-treated patients than control patients, which suggests that ALA-treated grafts are less inflammatory than untreated grafts. These results showed that ALA is safe for use in LT, induces gene changes that protect against hypoxia and oxidative stress and reduces the appearance of PRS., (© 2018 Steunstichting ESOT.)
- Published
- 2018
- Full Text
- View/download PDF
46. Amphipathic helical peptides hamper protein-protein interactions of the intrinsically disordered chromatin nuclear protein 1 (NUPR1).
- Author
-
Santofimia-Castaño P, Rizzuti B, Abián O, Velázquez-Campoy A, Iovanna JL, and Neira JL
- Subjects
- Basic Helix-Loop-Helix Transcription Factors genetics, Humans, Hydrophobic and Hydrophilic Interactions, Molecular Dynamics Simulation, Mutation, Neoplasm Proteins genetics, Protein Binding, Protein Conformation, Protein Interaction Domains and Motifs, Basic Helix-Loop-Helix Transcription Factors chemistry, Basic Helix-Loop-Helix Transcription Factors metabolism, Capsid Proteins chemistry, Neoplasm Proteins chemistry, Neoplasm Proteins metabolism, Peptide Fragments chemistry, Peptide Fragments metabolism
- Abstract
Background: NUPR1 is a multifunctional intrinsically disordered protein (IDP) involved, among other functions, in chromatin remodelling, and development of pancreatic ductal adenocarcinoma (PDAC). It interacts with several biomolecules through hydrophobic patches around residues Ala33 and Thr68. The drug trifluoperazine (TFP), which hampers PDAC development in xenografted mice, also binds to those regions. Because of the large size of the hot-spot interface of NUPR1, small molecules could not be adequate to modulate its functions., Methods: We explored how amphipathic helical-designed peptides were capable of interacting with wild-type NUPR1 and the Thr68Gln mutant, inhibiting the interaction with NUPR1 protein partners. We used in vitro biophysical techniques (fluorescence, circular dichroism (CD), nuclear magnetic resonance (NMR) and isothermal titration calorimetry (ITC)), in silico studies (docking and molecular dynamics (MD)), and in cellulo protein ligation assays (PLAs) to study the interaction., Results: Peptide dissociation constants towards wild-type NUPR1 were ~ 3 μM, whereas no interaction was observed with the Thr68Gln mutant. Peptides interacted with wild-type NUPR1 residues around Ala33 and residues at the C terminus, as shown by NMR. The computational results clarified the main determinants of the interactions, providing a mechanism for the ligand-capture that explains why peptide binding was not observed for Thr68Gln mutant. Finally, the in cellulo assays indicated that two out of four peptides inhibited the interaction of NUPR1 with the C-terminal region of the Polycomb RING protein 1 (C-RING1B)., Conclusions: Designed peptides can be used as lead compounds to inhibit NUPR1 interactions., General Significance: Peptides may be exploited as drugs to target IDPs., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
47. Ebselen impairs cellular oxidative state and induces endoplasmic reticulum stress and activation of crucial mitogen-activated protein kinases in pancreatic tumour AR42J cells.
- Author
-
Santofimia-Castaño P, Izquierdo-Alvarez A, Plaza-Davila M, Martinez-Ruiz A, Fernandez-Bermejo M, Mateos-Rodriguez JM, Salido GM, and Gonzalez A
- Subjects
- Amylases metabolism, Animals, Calcium metabolism, Cell Line, Tumor, Cell Survival drug effects, Gene Expression Regulation, Neoplastic drug effects, Isoindoles, Pancreatic Neoplasms drug therapy, Phosphorylation, Rats, Signal Transduction drug effects, Azoles pharmacology, Endoplasmic Reticulum Stress drug effects, Mitogen-Activated Protein Kinases metabolism, Organoselenium Compounds pharmacology, Oxidative Stress drug effects, Pancreatic Neoplasms metabolism
- Abstract
Ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one) is an organoselenium radical scavenger compound, which has strong antioxidant and anti-inflammatory effects. However, evidence suggests that this compound could exert deleterious actions on cell physiology. In this study, we have analyzed the effect of ebselen on rat pancreatic AR42J cells. Cytosolic free-Ca
2+ concentration ([Ca2+ ]c ), cellular oxidative status, setting of endoplasmic reticulum stress, and phosphorylation of major mitogen-activated protein kinases were analyzed. Our results show that ebselen evoked a concentration-dependent increase in [Ca2+ ]c . The compound induced an increase in the generation of reactive oxygen species in the mitochondria. We also observed an increase in global cysteine oxidation in the presence of ebselen. In the presence of ebselen an impairment of cholecystokinin-evoked amylase release was noted. Moreover, involvement of the unfolded protein response markers, ER chaperone and signaling regulator GRP78/BiP, eukaryotic translation initiation factor 2α and X-box binding protein 1 was detected. Finally, increases in the phosphorylation of SAPK/JNK, p38 MAPK, and p44/42 MAPK in the presence of ebselen were also observed. Our results provide evidences for an impairment of cellular oxidative state and enzyme secretion, the induction of endoplasmic reticulum stress and the activation of crucial mitogen-activated protein kinases in the presence of ebselen. As a consequence ebselen exerts a potential toxic effect on AR42J cells., (© 2017 Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
48. Intrinsically disordered chromatin protein NUPR1 binds to the C-terminal region of Polycomb RING1B.
- Author
-
Santofimia-Castaño P, Rizzuti B, Pey ÁL, Soubeyran P, Vidal M, Urrutia R, Iovanna JL, and Neira JL
- Abstract
Intrinsically disordered proteins (IDPs) are ubiquitous in eukaryotes, and they are often associated with diseases in humans. The protein NUPR1 is a multifunctional IDP involved in chromatin remodeling and in the development and progression of pancreatic cancer; however, the details of such functions are unknown. Polycomb proteins are involved in specific transcriptional cascades and gene silencing. One of the proteins of the Polycomb complex is the Ring finger protein 1 (RING1). RING1 is related to aggressive tumor features in multiple cancer types. In this work we characterized the interaction between NUPR1 and the paralogue RING1B in vitro, in silico, and in cellulo. The interaction occurred through the C-terminal region of RING1B (C-RING1B), with an affinity in the low micromolar range (∼10 μM). The binding region of NUPR1, mapped by NMR, was a hydrophobic polypeptide patch at the 30s region of its sequence, as pinpointed by computational results and site-directed mutagenesis at Ala33. The association between C-RING1B and wild-type NUPR1 also occurred in cellulo as tested by protein ligation assays; this interaction is inhibited by trifluoperazine, a drug known to hamper binding of wild-type NUPR1 with other proteins. Furthermore, the Thr68Gln and Ala33Gln/Thr68Gln mutants had a reduction in the binding toward C-RING1B as shown by in vitro, in silico, and in cellulo studies. This is an example of a well-folded partner of NUPR1, because its other interacting proteins are also unfolded. We hypothesize that NUPR1 plays an active role in chromatin remodeling and carcinogenesis, together with Polycomb proteins., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
49. Ebselen alters cellular oxidative status and induces endoplasmic reticulum stress in rat hippocampal astrocytes.
- Author
-
Santofimia-Castaño P, Izquierdo-Alvarez A, de la Casa-Resino I, Martinez-Ruiz A, Perez-Lopez M, Portilla JC, Salido GM, and Gonzalez A
- Subjects
- Animals, Astrocytes pathology, Azoles administration & dosage, Calcium metabolism, Cysteine metabolism, Dose-Response Relationship, Drug, Glial Fibrillary Acidic Protein metabolism, Glutathione Transferase metabolism, Hippocampus cytology, Isoindoles, Malondialdehyde metabolism, Mitochondria drug effects, Mitochondria metabolism, Mitogen-Activated Protein Kinases metabolism, Organoselenium Compounds administration & dosage, Oxidative Stress drug effects, Phosphorylation, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Antioxidants toxicity, Astrocytes drug effects, Azoles toxicity, Endoplasmic Reticulum Stress drug effects, Hippocampus drug effects, Organoselenium Compounds toxicity
- Abstract
Ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one) is an organoselenium radical scavenger compound, which has strong antioxidant and anti-inflammatory effects. Because of its properties, it may be protective against injury to the nervous tissue. However, evidence suggests that its glutathione peroxidase activity could underlie certain deleterious actions on cell physiology. In this study we have analyzed the effect of ebselen on rat hippocampal astrocytes in culture. Cellular oxidative status, cytosolic free-Ca(2+) concentration ([Ca(2+)]c), setting of endoplasmic reticulum stress and phosphorylation of glial fibrillary acidic protein and major mitogen-activated protein kinases were analyzed. Our results show that ebselen induced a concentration-dependent increase in the generation of reactive oxygen species in the mitochondria. We observed a concentration-dependent increase in global cysteine oxidation and in the level of malondialdehyde in the presence of ebselen. We also detected increases in catalase, glutathione S-transferase and glutathione reductase activity. Ebselen also evoked a concentration-dependent increase in [Ca(2+)]c. Moreover, we observed a concentration-dependent increase in the phosphorylation of the unfolded protein response markers, eukaryotic translation initiation factor 2α and X-box binding protein 1. Finally, ebselen also induced an increase in the phosphorylation of glial fibrillary acidic protein, SAPK/JNK, p38 MAPK and p44/42 MAPK. Our results provide strong evidence that implicate endoplasmic reticulum stress and activation of crucial mitogen-activated protein kinases in an oxidative damage of cells in the presence of ebselen. The compound thus might exert deleterious actions on astrocyte physiology that could compromise their function., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. Melatonin induces the expression of Nrf2-regulated antioxidant enzymes via PKC and Ca2+ influx activation in mouse pancreatic acinar cells.
- Author
-
Santofimia-Castaño P, Clea Ruy D, Garcia-Sanchez L, Jimenez-Blasco D, Fernandez-Bermejo M, Bolaños JP, Salido GM, and Gonzalez A
- Subjects
- Acinar Cells metabolism, Animals, Antioxidant Response Elements genetics, Calcium metabolism, Calcium Signaling genetics, Glutamate-Cysteine Ligase genetics, Glutamate-Cysteine Ligase metabolism, Heme Oxygenase-1 genetics, Heme Oxygenase-1 metabolism, Male, Mice, NAD(P)H Dehydrogenase (Quinone) genetics, NAD(P)H Dehydrogenase (Quinone) metabolism, NF-E2-Related Factor 2 genetics, Pancreas cytology, Phosphorylation, Protein Kinase C genetics, Antioxidants metabolism, Melatonin administration & dosage, NF-E2-Related Factor 2 biosynthesis, Pancreas metabolism, Protein Kinase C metabolism
- Abstract
The goal of this study was to evaluate the potential activation of the nuclear factor erythroid 2-related factor and the antioxidant-responsive element (Nrf2-ARE) signaling pathway in response to melatonin in isolated mouse pancreatic acinar cells. Changes in intracellular free Ca(2+) concentration were followed by fluorimetric analysis of fura-2-loaded cells. The activations of PKC and JNK were measured by Western blot analysis. Quantitative reverse transcription-polymerase chain reaction was employed to detect the expression of Nrf2-regulated antioxidant enzymes. Immunocytochemistry was employed to determine nuclear location of phosphorylated Nrf2, and the cellular redox state was monitored following MitoSOX Red-derived fluorescence. Our results show that stimulation of fura-2-loaded cells with melatonin (1 µM to 1 mM), in the presence of Ca(2+) in the extracellular medium, induced a slow and progressive increase of [Ca(2+)](c) toward a stable level. Melatonin did not inhibit the typical Ca(2+) response induced by CCK-8 (1 nM). When the cells were challenged with indoleamine in the absence of Ca(2+) in the extracellular solution (medium containing 0.5 mM EGTA) or in the presence of 1 mM LaCl(3), to inhibit Ca(2+) entry, we could not detect any change in [Ca(2+)](c). Nevertheless, CCK-8 (1 nM) was able to induce the typical mobilization of Ca(2+). When the cells were incubated with the PKC activator PMA (1 µM) in the presence of Ca(2+) in the extracellular medium, we observed a response similar to that noted when the cells were challenged with melatonin 100 µM. However, in the presence of Ro31-8220 (3 µM), a PKC inhibitor, stimulation of cells with melatonin failed to evoke changes in [Ca(2+)]c. Immunoblots, using an antibody specific for phospho-PKC, revealed that melatonin induces PKCα activation, either in the presence or in the absence of external Ca(2+). Melatonin induced the phosphorylation and nuclear translocation of the transcription factor Nrf2, and evoked a concentration-dependent increase in the expression of the antioxidant enzymes NAD(P)H-quinone oxidoreductase 1, catalytic subunit of glutamate-cysteine ligase, and heme oxygenase-1. Incubation of MitoSOX Red-loaded pancreatic acinar cells in the presence of 1 nM CCK-8 induced a statistically significant increase in dye-derived fluorescence, reflecting an increase in oxidation, that was abolished by pretreatment of cells with melatonin (100 µM) or PMA (1 µM). On the contrary, pretreatment with Ro31-8220 (3 µM) blocked the effect of melatonin on CCK-8-induced increase in oxidation. Finally, phosphorylation of JNK in the presence of CCK-8 or melatonin was also observed. We conclude that melatonin, via modulation of PKC and Ca(2+) signaling, could potentially stimulate the Nrf2-mediated antioxidant response in mouse pancreatic acinar cells., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.