José Luis Sierra Rodríguez
Complutense University of Madrid
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by José Luis Sierra Rodríguez.
Environmental Research | 2016
Xu Wang; M.A. Martínez; Menghong Dai; Dongmei Chen; Irma Ares; Alejandro Romero; Victor Castellano; Marta Martínez; José Luis Sierra Rodríguez; M.R. Martínez-Larrañaga; Arturo Anadón; Zonghui Yuan
Permethrin (PER), the most frequently used synthetic Type I pyrethroid insecticide, is widely used in the world because of its high activity as an insecticide and its low mammalian toxicity. It was originally believed that PER exhibited low toxicity on untargeted animals. However, as its use became more extensive worldwide, increasing evidence suggested that PER might have a variety of toxic effects on animals and humans alike, such as neurotoxicity, immunotoxicity, cardiotoxicity, hepatotoxicity, reproductive, genotoxic, and haematotoxic effects, digestive system toxicity, and cytotoxicity. A growing number of studies indicate that oxidative stress played critical roles in the various toxicities associated with PER. To date, almost no review has addressed the toxicity of PER correlated with oxidative stress. The focus of this article is primarily to summarise advances in the research associated with oxidative stress as a potential mechanism for PER-induced toxicity as well as its metabolism. This review summarises the research conducted over the past decade into the reactive oxygen species (ROS) generation and oxidative stress as a consequence of PER treatments, and ultimately their correlation with the toxicity and the metabolism of PER. The metabolism of PER involves various CYP450 enzymes, alcohol or aldehyde dehydrogenases for oxidation and the carboxylesterases for hydrolysis, through which oxidative stress might occur, and such metabolic factors are also reviewed. The protection of a variety of antioxidants against PER-induced toxicity is also discussed, in order to further understand the role of oxidative stress in PER-induced toxicity. This review will throw new light on the critical roles of oxidative stress in PER-induced toxicity, as well as on the blind spots that still exist in the understanding of PER metabolism, the cellular effects in terms of apoptosis and cell signaling pathways, and finally strategies to help to protect against its oxidative damage.
Environmental Research | 2016
José Luis Sierra Rodríguez; Irma Ares; Victor Castellano; M.A. Martínez; M.R. Martínez-Larrañaga; Arturo Anadón; M. Martínez
The effects of cyfluthrin oral exposure (1, 5, 10 and 20mg/kg bw, 6 days) on brain region monoamine levels of male rats were examined. Cyfluthrin-treated rats (1, 5 and 10mg/kg bw, orally 6 days), had no visible injury, i.e., no clinical signs of dysfunction were observed. However, rats treated with cyfluthrin at the highest dose (20mg/kg bw, orally 6 days) showed skeletal muscle contraction in the hind limbs, slight movement incoordination without any signs of dyskinesia and tremor after 1-2h of treatment. These signs were reversible at 6h after dose. After last dose of cyfluthrin, dopamine (DA) and serotonin (5-HT) and its metabolites levels were determined in brain regions hypothalamus, midbrain, hippocampus, striatum and prefrontal cortex by HPLC. Cyfluthrin (1mg/kg bw, orally 6 days) did not affect the DA, 5-HT and metabolites levels in the brain regions studied. Cyfluthrin (5, 10 and 20mg/kg bw, orally 6 days) caused a statistically significant decrease in DA and its metabolites DOPAC and HVA levels and in 5-HT and its metabolite 5-HIAA levels in a brain region- and dose-related manner. Moreover, cyfluthrin (20mg/kg bw, orally 6 days) evoked a statistically significant increase in 5-HT turnover in striatum and midbrain, and in DA turnover in striatum and prefrontal cortex. These findings indicate that serotoninergic and dopaminergic neurotransmission is affected by exposure to cyfluthrin and may contribute to the overall spectrum of neurotoxicity caused by this pyrethroid.
Revista Iberoamericana De Tecnologías Del Aprendizaje | 2014
Francisco-José García-Peñalvo; Antonio Sarasa Cabezuelo; José Luis Sierra Rodríguez
Innovation and engineering are very close concepts. Innovation is one of the key competences of the engineers in the way they use their own creativity and knowledge base to face the problems they have to resolve for humanity’s improvement and social evolution. In this special section, we have selected four papers from three research events (CINAIC 2013, TEEM 2013, and ISELEAR 2013) that empower the innovation and research cycles in engineering from different perspectives.
Environmental Research | 2018
M.A. Martínez; Irma Ares; José Luis Sierra Rodríguez; Marta Martínez; M.R. Martínez-Larrañaga; Arturo Anadón
ABSTRACT The effects of glyphosate oral exposure (35, 75, 150 and 800 mg/kg bw, 6 days) on brain region monoamine levels of male Wistar rats were examined. Glyphosate‐treated rats (35, 75, 150 and 800 mg/kg bw, 6 days), had no visible injury, i.e., no clinical signs of dysfunction were observed. After last dose of glyphosate, serotonin (5‐HT), dopamine (DA) and norepinephrine (NE) and its metabolites levels were determined in the brain regions striatum, hippocampus, prefrontal, cortex, hypothalamus and midbrain, by HPLC. Glyphosate caused statistically significant changes in the 5‐HT and its metabolite 5‐hydroxy‐3‐indolacetic acid (5‐HIAA), DA and its metabolites 3,4‐hydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), and NE and its metabolite 3‐metoxy‐4‐hydroxyphenylethyleneglycol (MHPG) levels in a brain regional‐ and dose‐related manner. Moreover, glyphosate, dose‐dependent, evoked a statistically significant increase in 5‐HT turnover in striatum and hypothalamus and in DA turnover in prefrontal cortex and hippocampus, and a statistically significant decrease in NE turnover in prefrontal cortex and hypothalamus. The present findings indicate that glyphosate significantly altered central nervous system (CNS) monoaminergic neurotransmitters in a brain regional‐ and dose‐related manner, effects that may contribute to the overall spectrum of neurotoxicity caused by this herbicide. HIGHLIGHTSGlyphosate oral exposure caused neurotoxicity in rats.Brain regions were susceptible to changes in CNS monoamine levels.Glyphosate reduced 5‐HT, DA, NE levels in a brain regional‐ and dose‐related manner.Glyphosate altered the serotoninergic, dopaminergic and noradrenergic systems.
Drug Metabolism Reviews | 2017
Xu Wang; Qinghua Wu; Aimei Liu; Arturo Anadón; José Luis Sierra Rodríguez; M.R. Martínez-Larrañaga; Zonghui Yuan; M.A. Martínez
Abstract Paracetamol (APAP) is one of the most widely used and popular over-the-counter analgesic and antipyretic drugs in the world when used at therapeutic doses. APAP overdose can cause severe liver injury, liver necrosis and kidney damage in human beings and animals. Many studies indicate that oxidative stress is involved in the various toxicities associated with APAP, and various antioxidants were evaluated to investigate their protective roles against APAP-induced liver and kidney toxicities. To date, almost no review has addressed the APAP toxicity in relation to oxidative stress. This review updates the research conducted over the past decades into the production of reactive oxygen species (ROS), reactive nitrogen species (RNS), and oxidative stress as a result of APAP treatments, and ultimately their correlation with the toxicity and metabolism of APAP. The metabolism of APAP involves various CYP450 enzymes, through which oxidative stress might occur, and such metabolic factors are reviewed within. The therapeutics of a variety of compounds against APAP-induced organ damage based on their anti-oxidative effects is also discussed, in order to further understand the role of oxidative stress in APAP-induced toxicity. This review will throw new light on the critical roles of oxidative stress in APAP-induced toxicity, as well as on the contradictions and blind spots that still exist in the understanding of APAP toxicity, the cellular effects in terms of organ injury and cell signaling pathways, and finally strategies to help remedy such against oxidative damage.
Science of The Total Environment | 2018
M.A. Martínez; Irma Ares; José Luis Sierra Rodríguez; Marta Martínez; David Roura-Martínez; Victor Castellano; Bernardo Lopez-Torres; M.R. Martínez-Larrañaga; Arturo Anadón
This study aimed to examine in rats the effects of the Type II pyrethroid lambda-cyhalothrin on hepatic microsomal cytochrome P450 (CYP) isoform activities, oxidative stress markers, gene expression of proinflammatory, oxidative stress and apoptosis mediators, and CYP isoform gene expression and metabolism phase I enzyme PCR array analysis. Lambda-cyhalothrin, at oral doses of 1, 2, 4 and 8mg/kg bw for 6days, increased, in a dose-dependent manner, hepatic activities of ethoxyresorufin O-deethylase (CYP1A1), methoxyresorufin O-demethylase (CYP1A2), pentoxyresorufin O-depentylase (CYP2B1/2), testosterone 7α- (CYP2A1), 16β- (CYP2B1), and 6β-hydroxylase (CYP3A1/2), and lauric acid 11- and 12-hydroxylase (CYP4A1/2). Similarly, lambda-cyhalothrin (4 and 8mg/kg bw, for 6days), in a dose-dependent manner, increased significantly hepatic CYP1A1, 1A2, 2A1, 2B1, 2B2, 2E1, 3A1, 3A2 and 4A1 mRNA levels and IL-1β, NFκB, Nrf2, p53, caspase-3 and Bax gene expressions. PCR array analysis showed from 84 genes examined (P<0.05; fold change>1.5), changes in mRNA levels in 18 genes: 13 up-regulated and 5 down-regulated. A greater fold change reversion than 3-fold was observed on the up-regulated ALDH1A1, CYP2B2, CYP2C80 and CYP2D4 genes. Ingenuity Pathway Analysis (IPA) groups the expressed genes into biological mechanisms that are mainly related to drug metabolism. In the top canonical pathways, Oxidative ethanol degradation III together with Fatty Acid α-oxidation may be significant pathways for lambda-cyhalothrin. Our results may provide further understanding of molecular aspects involved in lambda-cyhalothrin-induced liver injury.
Pharmacology & Therapeutics | 2018
Aimei Liu; Qinghua Wu; Jingchao Guo; Irma Ares; José Luis Sierra Rodríguez; M.R. Martínez-Larrañaga; Zonghui Yuan; Arturo Anadón; Xu Wang; M.A. Martínez
Abstract Statins, 3‐hydroxy‐3‐methylglutaryl‐coenzyme A reductase inhibitors, are currently the most effective lipid‐lowering drugs, effectively reducing the plasma total cholesterol and low‐density lipoprotein, while also decreasing three triacylglycerols and increasing plasma high‐density lipoprotein to a certain extent. However, the excessive or long‐term use of statins can cause in vitro cytotoxicity, in vivo liver injury, liver necrosis, kidney damage, and myopathy in both human beings and animals. Many studies indicate that oxidative stress is involved in the various toxicities associated with statins, and various antioxidants have been evaluated to investigate their protective roles against statin‐induced liver, kidney, and muscle toxicities. Widespread attention has been given to statin‐induced oxidative stress, with and without the use of other drugs. Much of the information about the mechanism for this reduction comes from cell culture and in experimental animal studies. The primary focus of this article is to summarize the research progress associated with oxidative stress as a plausible mechanism for statin‐induced toxicity, as well as its metabolic interactions. This review summarizes the research conducted over the past five years into the production of reactive oxygen species, oxidative stress as a result of statin treatments, and their correlation with statin‐induced toxicity and metabolism. Statin‐induced metabolism involves various CYP450 enzymes, which provide potential sites for statin‐induced oxidative stress, and these metabolic factors are also reviewed. The therapeutics of a variety of compounds against statin‐induced organ damage based on their anti‐oxidative effects is also discussed to further understand the role of oxidative stress in statin‐induced toxicity. This review sheds new light on the critical roles of oxidative stress in statin‐induced toxicity and prevention of this oxidative damage, as well as on the contradictions and unknowns that still exist regarding statin toxicity and the cellular effects in terms of organ injury and cell signaling pathways.
Food and Chemical Toxicology | 2018
José Luis Sierra Rodríguez; Irma Ares; Marta Martínez; M.R. Martínez-Larrañaga; Arturo Anadón; M.A. Martínez
Toxicokinetics of cyfluthrin after single oral [20 mg/kg body weight (bw)] and intravenous (IV) (3 mg/kg bw) doses were studied in rats. Serial blood samples were obtained after oral and IV administration. Brain tissue samples were also collected after oral administration. Cyfluthrin concentrations in plasma and brain tissues (hypothalamus, striatum, hippocampus and frontal cortex) were quantified using liquid chromatography tandem mass spectrometry (LC/MS). Cyfluthrin disposition was best described by the use of a two-compartment open model. When given orally, plasma kinetics showed an extensive oral absorption of cyfluthrin and a slow elimination. The area under the concentration-time curve [AUC (0-24h)] and maximal plasma concentration (Cmax) were 6.11 ± 1.06 mg h/L and 0.385 ± 0.051 μg/mL, respectively; β phase elimination half-life (T1/2β) was (17.15 ± 1.67 h). Oral bioavailability was found to be 71.60 ± 12.36%. After oral administration, cyfluthrin was widely distributed to brain tissues. AUC (0-24h) was significant higher in all tested brain tissues than in plasma. The largest discrepancy was found for hypothalamus. AUC (0-24h), Cmax and T1/2β in hypothalamus were 19.36 ± 2.56 mg h/L, 1.21 ± 0.11 μg/g and 22.73 ± 1.60 h, respectively. Assuming the identified toxicokinetics parameters, this study serves to better understand mammalian toxicity of pyrethroid cyfluthrin and to design further studies to characterize its neurotoxicity.
Frontiers in Veterinary Science | 2017
M.A. Martínez; Irma Ares; José Luis Sierra Rodríguez; Marta Martínez; M.R. Martínez-Larrañaga; Gerardo Isea; Arturo Anadón
The pharmacokinetics of pefloxacin after single 10 mg/kg BW intravenous (IV) and oral doses were studied in healthy broiler chickens. For 24 h, serial blood samples were obtained after IV and oral administration. Concentrations of pefloxacin and its major metabolite N-demethyl pefloxacin (norfloxacin) were measured by use of high-performance liquid chromatography. The plasma concentrations–time data were found to fit a two-compartment open model. For pefloxacin, the elimination half-life (t½β) was 8.44 ± 0.48 and 13.18 ± 0.82 h after IV and oral administration, respectively. After single oral dose, pefloxacin was rapidly absorbed with an absorption half-life (t½a) and TMAX of 0.87 ± 0.07 and 2.01 ± 0.12 h, respectively. Maximum plasma concentration (CMAX) was 4.02 ± 0.31 µg/mL. Oral bioavailability of pefloxacin was found to be 70 ± 2%. Pefloxacin was converted to N-demethyl pefloxacin (norfloxacin). This metabolite represented 5% of the parent drug plasma concentrations. The maximal plasma concentration (CMAX) of N-demethyl pefloxacin (norfloxacin) was calculated as 0.19 ± 0.01 mg/mL. The t½β of N-demethyl pefloxacin after oral pefloxacin administration was 10.93 ± 0.80 h. The results indicate that an oral dose of 10 mg pefloxacin/kg BW, every 24 h, should be effective in treatment of the most systemic infections in poultry.
Archive | 2013
Juan Manuel Dodero; Francisco José García-Peñalvo; Carina Soledad González González; Pablo Moreno Ger; Miguel Ángel Redondo; Antonio Sarasa Cabezuelo; José Luis Sierra Rodríguez