Network


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

Hotspot


Dive into the research topics where Laura del Barrio is active.

Publication


Featured researches published by Laura del Barrio.


Neurochemistry International | 2012

Guanosine protects human neuroblastoma SH-SY5Y cells against mitochondrial oxidative stress by inducing heme oxigenase-1 via PI3K/Akt/GSK-3β pathway.

Tharine Dal-Cim; Simone Molz; Javier Egea; Esther Parada; Alejandro Romero; Josiane Budni; María Dolores Martín de Saavedra; Laura del Barrio; Carla I. Tasca; Manuela G. López

Mitochondrial perturbation and oxidative stress are key factors in neuronal vulnerability in several neurodegenerative diseases or during brain ischemia. Here we have investigated the protective mechanism of action of guanosine, the guanine nucleoside, in a human neuroblastoma cell line, SH-SY5Y, subjected to mitochondrial oxidative stress. Blockade of mitochondrial complexes I and V with rotenone plus oligomycin (Rot/oligo) caused a significant decrease in cell viability and an increase in ROS production. Guanosine that the protective effect of guanosine incubated concomitantly with Rot/oligo abolished Rot/oligo-induced cell death and ROS production in a concentration dependent manner; maximum protection was achieved at the concentration of 1mM. The cytoprotective effect afforded by guanosine was abolished by adenosine A(1) or A(2A) receptor antagonists (DPCPX or ZM241385, respectively), or by a large (big) conductance Ca(2+)-activated K(+) channel (BK) blocker (charybdotoxin). Evaluation of signaling pathways showed that the protective effect of guanosine was not abolished by a MEK inhibitor (PD98059), by a p38(MAPK) inhibitor (SB203580), or by a PKC inhibitor (cheleritrine). However, when blocking the PI3K/Akt pathway with LY294002, the neuroprotective effect of guanosine was abolished. Guanosine increased Akt and p-Ser-9-GSK-3β phosphorylation confirming this pathway plays a key role in guanosines neuroprotective effect. Guanosine induced the antioxidant enzyme heme oxygenase-1 (HO-1) expression. The protective effects of guanosine were prevented by heme oxygenase-1 inhibitor, SnPP. Moreover, bilirubin, an antioxidant and physiologic product of HO-1, is protective against mitochondrial oxidative stress. In conclusion, our results show that guanosine can afford protection against mitochondrial oxidative stress by a signaling pathway that implicates PI3K/Akt/GSK-3β proteins and induction of the antioxidant enzyme HO-1.


Free Radical Biology and Medicine | 2010

Poststress treatment with PNU282987 can rescue SH-SY5Y cells undergoing apoptosis via α7 nicotinic receptors linked to a Jak2/Akt/HO-1 signaling pathway.

Esther Parada; Javier Egea; Alejandro Romero; Laura del Barrio; Antonio G. García; Manuela G. López

Most neuroprotection studies with nicotinic agonists have shown efficacy when given before the stressor. Here we have investigated whether the α7 nicotinic acetylcholine receptor (nAChR) agonist PNU282987 can prevent cell death once the cells have already undergone an oxidative stress. The combination of rotenone (30 μM) plus oligomycin A (10 μM) (rot/oligo) has been used as an in vitro model of mitochondrial ROS production. SH-SY5Y cells incubated with rot/oligo for 8h and left for another 16 h in MEM/F-12 experienced 30% apoptotic cell death. Under these experimental conditions, PNU282987 administered after rot/oligo (PST/PNU) prevented cell death in a concentration-dependent manner. Co-incubation of PNU282987 with 100 nM methyllycaconitine (a selective α7 nAChR antagonist), 10 μM mecamylamine (a nonselective nAChR antagonist), 3 μM LY294002 (a PI3K inhibitor), or 10 μM AG490 (a Jak2 inhibitor) prevented the protection afforded by PST/PNU. Moreover, the increase in ROS, active caspase-3, and apoptosis caused by rot/oligo was also prevented by PST/PNU. Furthermore, PNU282987 increased the expression of heme oxygenase-1 (HO-1), a critical cell defense enzyme against oxidative stress; this increase was prevented by AG490 or LY294002. The HO-1 inhibitor Sn(IV) protoporphyrin-IX also inhibited the PST/PNU protecting effects. These results suggest that activation of α7 nAChR linked to the Jak2/PI3K/Akt cascade induces the antioxidant enzyme HO-1 to provide neuroprotection.


Psychoneuroendocrinology | 2013

Nrf2 participates in depressive disorders through an anti-inflammatory mechanism

María Dolores Martín-de-Saavedra; Josiane Budni; Mauricio P. Cunha; Vanessa Gómez-Rangel; Silvia Lorrio; Laura del Barrio; Isabel Lastres-Becker; Esther Parada; Rosa M. Tordera; Ana Lúcia S. Rodrigues; Antonio Cuadrado; Manuela G. López

A causative relationship between inflammation and depression is gradually gaining consistency. Because Nrf2 participates in inflammation, we hypothesized that Nrf2 could play a role in depressive disorders. In this study, we have observed that Nrf2 deletion in mice results in: (i) a depressive-like behavior evaluated as an increase in the immobility time in the tail-suspension test and by a decrease in the grooming time in the splash test, (ii) reduced levels of dopamine and serotonin and increased levels of glutamate in the prefrontal cortex, (iii) altered levels of proteins associated to depression such as VEGF and synaptophysin and (iv) microgliosis. Furthermore, treatment of Nrf2 knockout mice with the anti-inflammatory drug rofecoxib reversed their depressive-like behavior, while induction of Nrf2 by sulforaphane, in an inflammatory model of depression elicited by LPS, afforded antidepressant-like effects. In conclusion, our results indicate that chronic inflammation due to a deletion of Nrf2 can lead to a depressive-like phenotype while induction of Nrf2 could become a new and interesting target to develop novel antidepressive drugs.


Journal of Neuroscience Research | 2011

Neuroprotective effect of guanosine against glutamate‐induced cell death in rat hippocampal slices is mediated by the phosphatidylinositol‐3 kinase/Akt/ glycogen synthase kinase 3β pathway activation and inducible nitric oxide synthase inhibition

Simone Molz; Tharine Dal-Cim; Josiane Budni; María Dolores Martín-de-Saavedra; Javier Egea; Alejandro Romero; Laura del Barrio; Ana Lúcia S. Rodrigues; Manuela G. López; Carla I. Tasca

Excitotoxicity and cell death induced by glutamate are involved in many neurodegenerative disorders. We have previously demonstrated that excitotoxicity induced by millimolar concentrations of glutamate in hippocampal slices involves apoptotic features and glutamate‐induced glutamate release. Guanosine, an endogenous guanine nucleoside, prevents excitotoxicty by its ability to modulate glutamate transport. In this study, we have evaluated the neuroprotective effect of guanosine against glutamate‐induced toxicity in hippocampal slices and the mechanism involved in such an effect. We have found that guanosine (100 μM) was neuroprotective against 1 mM glutamate‐induced cell death through the inhibition of glutamate release induced by glutamate. Guanosine also induced the phosphorylation and, thus, activation of protein kinase B (PKB/Akt), a downstream target of phosphatidylinositol‐3 kinase (PI3K), as well as phosphorylation of glycogen synthase kinase 3β, which has been reported to be inactivated by Akt after phosphorylation at Ser9. Glutamate treated hippocampal slices showed increased inducible nitric oxide synthase (iNOS) expression that was prevented by guanosine. Slices preincubated with SNAP (an NO donor), inhibited the protective effect of guanosine. LY294002 (30 μM), a PI3K inhibitor, attenuated guanosine‐induced neuroprotection, guanosine prevention of glutamate release, and guanosine‐induced GSK3βSer9 phosphorylation but not guanosine reduction of glutamate‐induced iNOS expression. Taken together, the results of this study show that guanosine protects hippocampal slices by a mechanism that involves the PI3K/Akt/GSK3βSer9 pathway and prevention of glutamate‐induced glutamate release. Furthermore, guanosine also reduces glutamate‐induced iNOS by a PI3K/Akt‐independent mechanism.


European Journal of Medicinal Chemistry | 2011

N-Acylaminophenothiazines: neuroprotective agents displaying multifunctional activities for a potential treatment of Alzheimer’s disease

Gema C. González-Muñoz; Mariana P. Arce; Beatriz López; Concepción Pérez; Alejandro Romero; Laura del Barrio; María Dolores Martín-de-Saavedra; Javier Egea; Rafael León; Mercedes Villarroya; Manuela G. López; Antonio G. García; Santiago Conde; María Isabel Rodríguez-Franco

We have previously reported the multifunctional profile of N-(3-chloro-10H-phenothiazin-10-yl)-3-(dimethylamino)propanamide (1) as an effective neuroprotectant and selective butyrylcholinesterase inhibitor. In this paper, we have developed a series of N-acylaminophenothiazines obtained from our compound library or newly synthesised. At micro- and sub-micromolar concentrations, these compounds selectively inhibited butyrylcholinesterase (BuChE), protected neurons against damage caused by both exogenous and mitochondrial free radicals, showed low toxicity, and could penetrate into the CNS. In addition, N-(3-chloro-10H-phenothiazin-10-yl)-2-(pyrrolidin-1-yl)acetamide (11) modulated the cytosolic calcium concentration and protected human neuroblastoma cells against several toxics, such as calcium overload induced by an L-type Ca2+-channel agonist, tau-hyperphosphorylation induced by okadaic acid and Aβ peptide.


Chemico-Biological Interactions | 2010

Peptide gomesin triggers cell death through L-type channel calcium influx, MAPK/ERK, PKC and PI3K signaling and generation of reactive oxygen species.

Rossana C. Soletti; Laura del Barrio; Sirlei Daffre; Antonio Miranda; Helena L. Borges; Vivaldo Moura-Neto; Manuela G. López; Nelson H. Gabilan

Gomesin is an antimicrobial peptide isolated from hemocytes of a common Brazilian tarantula spider named Acanthoscurria gomesiana. This peptide exerts antitumor activity in vitro and in vivo by an unknown mechanism. In this study, the cytotoxic mechanism of gomesin in human neuroblastoma SH-SY5Y and rat pheochromocytoma PC12 cells was investigated. Gomesin induced necrotic cell death and was cytotoxic to SH-SY5Y and PC12 cells. The peptide evoked a rapid and transient elevation of intracellular calcium levels in Fluo-4-AM loaded PC12 cells, which was inhibited by nimodipine, an L-type calcium channel blocker. Preincubation with nimodipine also inhibited cell death induced by gomesin in SH-SY5Y and PC12 cells. Gomesin-induced cell death was prevented by the pretreatment with MAPK/ERK, PKC or PI3K inhibitors, but not with PKA inhibitor. In addition, gomesin generated reactive oxygen species (ROS) in SH-SY5Y cells, which were blocked with nimodipine and MAPK/ERK, PKC or PI3K inhibitors. Taken together, these results suggest that gomesin could be a useful anticancer agent, which mechanism of cytotoxicity implicates calcium entry through L-type calcium channels, activation of MAPK/ERK, PKC and PI3K signaling as well as the generation of reactive oxygen species.


Bioorganic & Medicinal Chemistry | 2011

Cholinergic and neuroprotective drugs for the treatment of Alzheimer and neuronal vascular diseases. II. Synthesis, biological assessment, and molecular modelling of new tacrine analogues from highly substituted 2-aminopyridine-3-carbonitriles.

Abdelouahid Samadi; Carolina Valderas; Cristóbal de los Ríos; Agatha Bastida; Mourad Chioua; Laura González-Lafuente; Inés Colmena; Luis Gandía; Alejandro Romero; Laura del Barrio; María Dolores Martín-de-Saavedra; Manuela G. López; Mercedes Villarroya; José Marco-Contelles

The synthesis, biological assessment, and molecular modelling of new tacrine analogues 11-22 is described. Compounds 11-22 have been obtained by Friedländer-type reaction of 2-aminopyridine-3-carbonitriles 1-10 with cyclohexanone or 1-benzyl-4-piperidone. The biological evaluation showed that some of these molecules were good AChE inhibitors, in the nanomolar range, and quite selective regarding the inhibition of BuChE, the most potent being 5-amino-2-(dimethylamino)-6,7,8,9-tetrahydrobenzo[1,8-b]-naphthyridine-3-carbonitrile (11) [IC(50) (EeAChE: 14nM); IC(50) (eqBuChE: 5.2μM]. Kinetic studies on the easily available and potent anticholinesterasic compound 5-amino-2-(methoxy)-6,7,8,9-tetrahydrobenzo[1,8-b]-naphthyridine-3-carbonitrile (16) [IC(50) (EeAChE: 64nM); IC(50) (eqBuChE: 9.6μM] showed that this compound is a mixed-type inhibitor (K(i)=69.2nM) of EeAChE. Molecular modelling on inhibitor 16 confirms that this compound, as expected and similarly to tacrine, binds at the catalytic active site of EeAChE. The neuroprotective profile of molecules 11-22 has been investigated in SH-SY5Y neuroblastoma cells stressed with a mixture of oligomycin-A/rotenone. Compound 16 was also able to rescue by 50% cell death induced by okadaic acid in SH-SY5Y cells. From these results we conclude that the neuroprotective profile of these molecules is moderate, the most potent being compounds 12 and 17 which reduced cell death by 29%. Compound 16 does not affect ACh- nor K(+)-induced calcium signals in bovine chromaffin cells. Consequently, tacrine analogues 11-22 can be considered attractive therapeutic molecules on two key pharmacological targets playing key roles in the progression of Alzheimer, that is, cholinergic dysfunction and oxidative stress, as well as in neuronal cerebrovascular diseases.


British Journal of Pharmacology | 2011

Calcium signalling mediated through α7 and non-α7 nAChR stimulation is differentially regulated in bovine chromaffin cells to induce catecholamine release

Laura del Barrio; Javier Egea; Rafael León; Alejandro Romero; Ana Isabel Ruiz; Mayte Montero; Javier Alvarez; Manuela G. López

BACKGROUND AND PURPOSE Ca2+ signalling and exocytosis mediated by nicotinic receptor (nAChR) subtypes, especially the α7 nAChR, in bovine chromaffin cells are still matters of debate.


Neurochemistry International | 2011

Chondroitin sulfate reduces cell death of rat hippocampal slices subjected to oxygen and glucose deprivation by inhibiting p38, NFκB and iNOS.

María Dolores Martín-de-Saavedra; Laura del Barrio; Noelia Cañas; Javier Egea; Silvia Lorrio; E. Montell; J. Vergés; Antonio G. García; Manuela G. López

The glycosaminoglycan chondroitin sulfate (CS) is a major constituent of the extracellular matrix of the central nervous system where it can constitute part of the perineuronal nets. Constituents of the perineuronal nets are gaining interest because they have modulatory actions on their neighbouring neurons. In this study we have investigated if CS could afford protection in an acute in vitro ischemia/reoxygenation model by using isolated hippocampal slices subjected to 60min oxygen and glucose deprivation (OGD) followed by 120min reoxygenation (OGD/Reox). In this toxicity model, CS afforded protection of rat hippocampal slices measured as a reduction of lactate dehydrogenase (LDH) release; maximum protection (70% reduction of LDH) was obtained at the concentration of 3mM. To evaluate the intracellular signaling pathways implicated in the protective effect of CS, we first analysed the participation of the mitogen-activated protein kinases (MAPKs) p38 and ERK1/2 by western blot. OGD/Reox induced the phosphorylation of p38 and dephosphorylation of ERK1/2; however, CS only inhibited p38 but had no effect on ERK1/2. Furthermore, OGD/Reox-induced translocation of p65 to the nucleus was prevented in CS treated hippocampal slices. Finally, CS inhibited iNOS induction caused by OGD/Reox and thereby nitric oxide (NO) production measured as a reduction in DAF-2 DA fluorescence. In conclusion, the protective effect of CS in hippocampal slices subjected to OGD/Reox can be related to a modulatory action of the local immune response by a mechanism that implies inhibition of p38, NFκB, iNOS and the production of NO.


Pain | 2009

Haeme oxygenase-1 overexpression via nAChRs and the transcription factor Nrf2 has antinociceptive effects in the formalin test

Javier Egea; Angelo O. Rosa; Silvia Lorrio; Laura del Barrio; Antonio Cuadrado; Manuela G. López

ABSTRACT Epibatidine has shown antinociceptive effects in various pain models, being 200‐fold more potent than morphine. Previous results from our laboratory demonstrated that HO‐1 overexpression has an antinociceptive effect in the formalin test. Furthermore, epibatidine was able to induce haeme oxygenase‐1 (HO‐1). So, the aim of this study was to investigate the effect of HO‐1 overexpression induced by epibatidine in nociception elicited by formalin injection in the mice hindpaw. Administration of epibatidine (4 μg/kg) 24 h before the test reduced the nociceptive response during the first phase and second phase of the formalin test. This effect was prevented by treatment with tin protoporphyrin (SnPP, an inhibitor of HO‐1 activity) administered via intraplantar 5 min before the test, suggesting a main role of HO‐1. Western blot analysis revealed that epibatidine treatment increased by 2‐fold HO‐1 expression in the paw; this effect was lost in knockout mice for nuclear factor‐erythroid 2‐related factor 2 (Nrf2) and was accompanied by the loss of its antinociceptive effect. Furthermore, the antinociceptive effect of epibatidine was related to the activation of alpha7 and/or alpha9 nAChRs since methyllycaconitine (MLA) and mecamylamine but not dihydro‐&bgr;‐erythroidine (DH&bgr;E) reverted this effect. Finally, we showed by flow cytometry and by immunofluorescence that white blood cells of the animals injected with epibatidine expressed more HO‐1 than control animals, and this expression was also reverted by MLA pre‐treatment. These findings demonstrate that HO‐1 induction by epibatidine has antinociceptive and anti‐inflammatory effects by the activation of MLA‐sensitive nAChRs.

Collaboration


Dive into the Laura del Barrio's collaboration.

Top Co-Authors

Avatar

Manuela G. López

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Javier Egea

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Alejandro Romero

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Antonio G. García

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mercedes Villarroya

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Esther Parada

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Rafael León

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Silvia Lorrio

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Antonio Cuadrado

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge