Network


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

Hotspot


Dive into the research topics where Sergio Rius-Pérez is active.

Publication


Featured researches published by Sergio Rius-Pérez.


Free Radical Biology and Medicine | 2017

Redox signaling in the gastrointestinal tract

Salvador Pérez; Raquel Taléns-Visconti; Sergio Rius-Pérez; Isabela Finamor; Juan Sastre

Redox signaling regulates physiological self-renewal, proliferation, migration and differentiation in gastrointestinal epithelium by modulating Wnt/β-catenin and Notch signaling pathways mainly through NADPH oxidases (NOXs). In the intestine, intracellular and extracellular thiol redox status modulates the proliferative potential of epithelial cells. Furthermore, commensal bacteria contribute to intestine epithelial homeostasis through NOX1- and dual oxidase 2-derived reactive oxygen species (ROS). The loss of redox homeostasis is involved in the pathogenesis and development of a wide diversity of gastrointestinal disorders, such as Barretts esophagus, esophageal adenocarcinoma, peptic ulcer, gastric cancer, ischemic intestinal injury, celiac disease, inflammatory bowel disease and colorectal cancer. The overproduction of superoxide anion together with inactivation of superoxide dismutase are involved in the pathogenesis of Barretts esophagus and its transformation to adenocarcinoma. In Helicobacter pylori-induced peptic ulcer, oxidative stress derived from the leukocyte infiltrate and NOX1 aggravates mucosal damage, especially in HspB+ strains that downregulate Nrf2. In celiac disease, oxidative stress mediates most of the cytotoxic effects induced by gluten peptides and increases transglutaminase levels, whereas nitrosative stress contributes to the impairment of tight junctions. Progression of inflammatory bowel disease relies on the balance between pro-inflammatory redox-sensitive pathways, such as NLRP3 inflammasome and NF-κB, and the adaptive up-regulation of Mn superoxide dismutase and glutathione peroxidase 2. In colorectal cancer, redox signaling exhibits two Janus faces: On the one hand, NOX1 up-regulation and derived hydrogen peroxide enhance Wnt/β-catenin and Notch proliferating pathways; on the other hand, ROS may disrupt tumor progression through different pro-apoptotic mechanisms. In conclusion, redox signaling plays a critical role in the physiology and pathophysiology of gastrointestinal tract.


Redox biology | 2017

Chronic aspartame intake causes changes in the trans-sulphuration pathway, glutathione depletion and liver damage in mice

Isabela Finamor; Salvador Pérez; Caroline A. Bressan; Carlos E. Brenner; Sergio Rius-Pérez; Patricia C. Brittes; Gabriele Cheiran; Maria Izabel de Ugalde Marques da Rocha; Marcelo da Veiga; Juan Sastre; Maria A. Pavanato

No-caloric sweeteners, such as aspartame, are widely used in various food and beverages to prevent the increasing rates of obesity and diabetes mellitus, acting as tools in helping control caloric intake. Aspartame is metabolized to phenylalanine, aspartic acid, and methanol. Our aim was to study the effect of chronic administration of aspartame on glutathione redox status and on the trans-sulphuration pathway in mouse liver. Mice were divided into three groups: control; treated daily with aspartame for 90 days; and treated with aspartame plus N-acetylcysteine (NAC). Chronic administration of aspartame increased plasma alanine aminotransferase (ALT) and aspartate aminotransferase activities and caused liver injury as well as marked decreased hepatic levels of reduced glutathione (GSH), oxidized glutathione (GSSG), γ-glutamylcysteine (γ-GC), and most metabolites of the trans-sulphuration pathway, such as cysteine, S-adenosylmethionine (SAM), and S-adenosylhomocysteine (SAH). Aspartame also triggered a decrease in mRNA and protein levels of the catalytic subunit of glutamate cysteine ligase (GCLc) and cystathionine γ-lyase, and in protein levels of methionine adenosyltransferase 1A and 2A. N-acetylcysteine prevented the aspartame-induced liver injury and the increase in plasma ALT activity as well as the decrease in GSH, γ-GC, cysteine, SAM and SAH levels and GCLc protein levels. In conclusion, chronic administration of aspartame caused marked hepatic GSH depletion, which should be ascribed to GCLc down-regulation and decreased cysteine levels. Aspartame triggered blockade of the trans-sulphuration pathway at two steps, cystathionine γ-lyase and methionine adenosyltransferases. NAC restored glutathione levels as well as the impairment of the trans-sulphuration pathway.


PLOS ONE | 2017

p38α regulates actin cytoskeleton and cytokinesis in hepatocytes during development and aging.

Ana M. Tormos; Sergio Rius-Pérez; María Jorques; Lorena Ramírez; Ángela M. Valverde; Ángel R. Nebreda; Juan Sastre; Raquel Taléns-Visconti

Background Hepatocyte poliploidization is an age-dependent process, being cytokinesis failure the main mechanism of polyploid hepatocyte formation. Our aim was to study the role of p38α MAPK in the regulation of actin cytoskeleton and cytokinesis in hepatocytes during development and aging. Methods Wild type and p38α liver-specific knock out mice at different ages (after weaning, adults and old) were used. Results We show that p38α MAPK deficiency induces actin disassembly upon aging and also cytokinesis failure leading to enhanced binucleation. Although the steady state levels of cyclin D1 in wild type and p38α knock out old livers remained unaffected, cyclin B1- a marker for G2/M transition- was significantly overexpressed in p38α knock out mice. Our findings suggest that hepatocytes do enter into S phase but they do not complete cell division upon p38α deficiency leading to cytokinesis failure and binucleation. Moreover, old liver-specific p38α MAPK knock out mice exhibited reduced F-actin polymerization and a dramatic loss of actin cytoskeleton. This was associated with abnormal hyperactivation of RhoA and Cdc42 GTPases. Long-term p38α deficiency drives to inactivation of HSP27, which seems to account for the impairment in actin cytoskeleton as Hsp27-silencing decreased the number and length of actin filaments in isolated hepatocytes. Conclusions p38α MAPK is essential for actin dynamics with age in hepatocytes.


The Journal of Pathology | 2018

Obesity causes Pgc-1α deficiency in the pancreas leading to marked Il-6 up-regulation via NF-κB in acute pancreatitis: PGC-1α deficiency up-regulates IL-6 via NF-κB in pancreatitis

Salvador Pérez; Sergio Rius-Pérez; Isabela Finamor; Pablo Martí-Andrés; Ignacio Prieto; Raquel Garcia; María Monsalve; Juan Sastre

Obesity is associated with local and systemic complications in acute pancreatitis. PPARγ coactivator 1α (PGC‐1α) is a transcriptional coactivator and master regulator of mitochondrial biogenesis that exhibits dysregulation in obese subjects. Our aims were: (1) to study PGC‐1α levels in pancreas from lean or obese rats and mice with acute pancreatitis; and (2) to determine the role of PGC‐1α in the inflammatory response during acute pancreatitis elucidating the signaling pathways regulated by PGC‐1α. Lean and obese Zucker rats and lean and obese C57BL6 mice were used first; subsequently, wild‐type and PGC‐1α knockout (KO) mice with cerulein‐induced pancreatitis were used to assess the inflammatory response and expression of target genes. Ppargc1a mRNA and protein levels were markedly downregulated in pancreas of obese rats and mice versus lean animals. PGC‐1α protein levels increased in pancreas of lean mice with acute pancreatitis, but not in obese mice with pancreatitis. Interleukin‐6 (Il6) mRNA levels were dramatically upregulated in pancreas of PGC‐1α KO mice after cerulein‐induced pancreatitis in comparison with wild‐type mice with pancreatitis. Edema and the inflammatory infiltrate were more intense in pancreas from PGC‐1α KO mice than in wild‐type mice. The lack of PGC‐1α markedly enhanced nuclear translocation of phospho‐p65 and recruitment of p65 to Il6 promoter. PGC‐1α bound phospho‐p65 in pancreas during pancreatitis in wild‐type mice. Glutathione depletion in cerulein‐induced pancreatitis was more severe in KO mice than in wild‐type mice. PGC‐1α KO mice with pancreatitis, but not wild‐type mice, exhibited increased myeloperoxidase activity in the lungs, together with alveolar wall thickening and collapse, which were abrogated by blockade of the IL‐6 receptor glycoprotein 130 with LMT‐28. In conclusion, obese rodents exhibit PGC‐1α deficiency in the pancreas. PGC‐1α acts as selective repressor of nuclear factor‐κB (NF‐κB) towards IL‐6 in pancreas. PGC‐1α deficiency markedly enhanced NF‐κB‐mediated upregulation of Il6 in pancreas in pancreatitis, leading to a severe inflammatory response. Copyright


Redox biology | 2018

Age-dependent regulation of antioxidant genes by p38α MAPK in the liver

Salvador Pérez; Sergio Rius-Pérez; Ana M. Tormos; Isabela Finamor; Angel R. Nebreda; Raquel Taléns-Visconti; Juan Sastre

p38α is a redox sensitive MAPK activated by pro-inflammatory cytokines and environmental, genotoxic and endoplasmic reticulum stresses. The aim of this work was to assess whether p38α controls the antioxidant defense in the liver, and if so, to elucidate the mechanism(s) involved and the age-related changes. For this purpose, we used liver-specific p38α-deficient mice at two different ages: young-mice (4 months-old) and old-mice (24 months-old). The liver of young p38α knock-out mice exhibited a decrease in GSH levels and an increase in GSSG/GSH ratio and malondialdehyde levels. However, old mice deficient in p38α had higher hepatic GSH levels and lower GSSG/GSH ratio than young p38α knock-out mice. Liver-specific p38α deficiency triggered a dramatic down-regulation of the mRNAs of the key antioxidant enzymes glutamate cysteine ligase, superoxide dismutase 1, superoxide dismutase 2, and catalase in young mice, which seems mediated by the lack of p65 recruitment to their promoters. Nrf-2 nuclear levels did not change significantly in the liver of young mice upon p38α deficiency, but nuclear levels of phospho-p65 and PGC-1α decreased in these mice. p38α-dependent activation of NF-κB seems to occur through classical IκB Kinase and via ribosomal S6 kinase1 and AKT in young mice. However, unexpectedly the long-term deficiency in p38α triggers a compensatory up-regulation of antioxidant enzymes via NF-κB activation and recruitment of p65 to their promoters. In conclusion, p38α MAPK maintains the expression of antioxidant genes in liver of young animals via NF-κΒ under basal conditions, whereas its long-term deficiency triggers compensatory up-regulation of antioxidant enzymes through NF-κΒ.


Neurologia | 2017

Vascular pathology: Cause or effect in Alzheimer disease?

Sergio Rius-Pérez; Ana M. Tormos; Salvador Pérez; Raquel Taléns-Visconti


Neurologia | 2015

Patología vascular: ¿causa o efecto en la enfermedad de Alzheimer?

Sergio Rius-Pérez; Ana M. Tormos; Salvador Pérez; Raquel Taléns-Visconti


Free Radical Biology and Medicine | 2018

Thioredoxin-related protein of 14 kDa may directly reduce protein cysteinylation motifs

Pablo Martí-Andrés; Isabela A. Finamor; Belén Espinosa; Salvador Pérez; Sergio Rius-Pérez; Raquel Taléns-Visconti; Rafel León; Antonio Martínez-Ruiz; Elias S.J. Arnér; Juan Sastre


Free Radical Biology and Medicine | 2018

Time-course of thiol oxidation of protein phosphatases during cerulein-induced acute pancreatitis

Sergio Rius-Pérez; Salvador Pérez; Pablo Martí-Andrés; Isabela Finamor; Raquel Taléns-Visconti; Antonio Cuadrado; Michel B. Toledano; Juan Sastre


Free Radical Biology and Medicine | 2017

p38α and NF-κB regulate antioxidant defense in the liver through an age-dependent mechanism

Salvador Pérez; Sergio Rius-Pérez; Isabela Finamor; Ana Isabel Martínez; Pablo Marti; Angel R. Nebreda; Raquel Taléns; Juan Sastre

Collaboration


Dive into the Sergio Rius-Pérez's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juan Sastre

University of Valencia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonio Cuadrado

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge