Person:
Pino Sans, Javier Del

Loading...
Profile Picture
First Name
Javier Del
Last Name
Pino Sans
Affiliation
Universidad Complutense de Madrid
Faculty / Institute
Veterinaria
Department
Farmacología y Toxicología
Area
Toxicología
Identifiers
UCM identifierORCIDScopus Author IDWeb of Science ResearcherIDDialnet IDGoogle Scholar ID

Search Results

Now showing 1 - 10 of 26
  • Item
    Guía de prácticas y seminarios de Fundamentos de Toxicología
    (2021) Ramos Alonso, Eva; Pino Sans, Javier Del; Romero Martínez, Manuel Alejandro; Díaz Plaza, María Jesús; Frejo Moya, María Teresa; Moyano-Cires Ivanoff, Paula Viviana
  • Item
    Primary hippocampal neuronal cell death induction after acute and repeated paraquat exposures mediated by AChE variants alteration and cholinergic and glutamatergic transmission disruption
    (Toxicology, 2017) Pino Sans, Javier Del; Moyano-Cires Ivanoff, Paula Viviana; Gómez Díaz, Gloria; Anadón Baselga, María José; Díaz Plaza, María Jesús; García Sánchez, José Manuel; Lobo Alonso, Margarita; Pelayo Alarcón, Adela; Sola Vendrell, Emma; Frejo Moya, María Teresa
    Paraquat (PQ) is a widely used non-selective contact herbicide shown to produce memory and learning deficits after acute and repeated exposure similar to those induced in Alzheimer's disease (AD). However, the complete mechanisms through which it induces these effects are unknown. On the other hand, cholinergic and glutamatergic systems, mainly in the hippocampus, are involved on learning, memory and cell viability regulation. An alteration of hippocampal cholinergic or glutamatergic transmissions or neuronal cell loss may induce these effects. In this regard, it has been suggested that PQ may induce cell death and affect cholinergic and glutamatergic transmission, which alteration could produce neuronal loss. According to these data, we hypothesized that PQ could induce hippocampal neuronal loss through cholinergic and glutamatergic transmissions alteration. To prove this hypothesis, we evaluated in hippocampal primary cell culture, the PQ toxic effects after 24h and 14 consecutive days exposure on neuronal viability and the cholinergic and glutamatergic mechanisms related to it. This study shows that PQ impaired acetylcholine levels and induced AChE inhibition and increased CHT expression only after 14days exposure, which suggests that acetylcholine levels alteration could be mediated by these actions. PQ also disrupted glutamate levels through induction of glutaminase activity. In addition, PQ induced, after 24h and 14days exposure, cell death on hippocampal neurons that was partially mediated by AChE variants alteration and cholinergic and gultamatergic transmissions disruption. Our present results provide new view of the mechanisms contributing to PQ neurotoxicity and may explain cognitive dysfunctions observed after PQ exposure.
  • Item
    Efectos del amitraz sobre neurotransmisores monoaminérgicos en el sistema nervioso central de rata
    (2010) Pino Sans, Javier Del; Martínez-Larrañaga, María Rosa; Anadón Navarro, Arturo
    Por todo ello, el presente trabajo de investigación tiene dos objetivos: (1) el estudio de las posibles alteraciones en los sistemas serotoninérgico, noradrenérgico y dopaminérgico en regiones del SNC de ratas de edad 30 y 60 días expuestas a dosis múltiples de amitraz, y (2) el estudio de las posibles alteraciones permanentes heredadas en ratas a la edad de 60 días, procedentes de madres a las que se les administró amitraz durante el periodo de la preñez y la lactancia. La elección de la rata como modelo animal experimental se justifica, en general, en que los roedores poseen una serie de ventajas para los estudios toxicológicos, como son su pequeño tamaño, fácil manejo, resistencia a infecciones, corto ciclo de vida y de gestación, y grandes camadas (en número). Además, es un modelo animal aceptado para proporcionar datos que pueden ser incorporados a la evaluación del riesgo para el hombre de pesticidas. En los estudios de niveles de neurotransmisores en el SNC, la rata tiene la ventaja añadida de que por el tamaño de su encéfalo permite una buena localización de las distintas regiones encefálicas. Además, a fecha de hoy existen distintos atlas que detallan la anatomía de su sistema nervioso y la distribución de las sustancias neuroactivas (PAROXINOS y WATSON, 1998; TOHYAMA y TAKATSUJI, 1998).
  • Item
    Cadmium alters heat shock protein pathways in SN56 cholinergic neurons, leading to AB and phosphorylated Tau protein generation and cell death
    (Food and Chemical Toxicology, 2018) Moyano-Cires Ivanoff, Paula Viviana; García Sánchez, José Manuel; Lobo Alonso, Margarita; Anadón Baselga, María José; Sola Vendrell, Emma; Pelayo Alarcón, Adela; García Lobo, Jimena; Frejo Moya, María Teresa; Pino Sans, Javier Del
    Cadmium, a neurotoxic environmental compound, produces cognitive disorders, although the mechanism remains unknown. Cadmium induces a more pronounced cell death on cholinergic neurons from basal forebrain (BF), mediated, in part, by increase in Aβ and total and phosphorylated Tau protein levels, which may explain cadmium effects on learning and memory processes. Cadmium downregulates the expression of heat shock proteins (HSPs) HSP 90, HSP70 and HSP27, and of HSF1, the master regulator of the HSP pathway. HSPs proteins reduce the production of Aβ and phosphorylated Tau proteins and avoid cell death pathways induction. Thus, we hypothesized that cadmium induced the production of Aβ and Tau proteins by HSP pathway disruption through HSF1 expression alteration, leading to BF cholinergic neurons cell death. Our results show that cadmium downregulates HSF1, leading to HSP90, HSP70 and HSP27 gene expression downregulation in BF SN56 cholinergic neurons. In addition, cadmium induced Aβ and total and phosphorylated Tau proteins generation, mediated partially by HSP90, HSP70 and HSP27 disruption, leading to cell death. These results provide new understanding of the mechanisms contributing to cadmium harmful effects on cholinergic neurons.
  • Item
    Neuroprotective Action of Multitarget 7-Aminophenanthridin-6(5H)-one Derivatives against Metal-Induced Cell Death and Oxidative Stress in SN56 Cells
    (ACS Chemical Neuroscience, 2021) Moyano-Cires Ivanoff, Paula Viviana; Vicente Zurdo, David; Blázquez-Barbadillo, Cristina; Menéndez Ramos, José Carlos; González Matilla, Juan Francisco; Rosales Conrado, Noelia; Pino Sans, Javier Del
    Neurodegenerative diseases have been associated with brain metal accumulation, which produces oxidative stress (OS), matrix metalloproteinases (MMPs) induction, and neuronal cell death. Several metals have been reported to downregulate both the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway and the antioxidant enzymes regulated by it, mediating OS induction and neurodegeneration. Among a recently discovered family of multitarget 7-amino-phenanthridin-6-one derivatives (APH) the most promising compounds were tested against metal-induced cell death and OS in SN56 cells. These compounds, designed to have chelating activity, are known to inhibit some MMPs and to present antioxidant and neuroprotective effects against hydrogen peroxide treatment to SN56 neuronal cells. However, the mechanisms that mediate this protective effect are not fully understood. The obtained results show that compounds APH1, APH2, APH3, APH4, and APH5 were only able to chelate iron and copper ions among all metals studied and that APH3, APH4, and APH5 were also able to chelate mercury ion. However, none of them was able to chelate zinc, cadmium, and aluminum, thus exhibiting selective chelating activity that can be partly responsible for their neuroprotective action. Otherwise, our results indicate that their antioxidant effect is mediated through induction of the Nrf2 pathway that leads to overexpression of antioxidant enzymes. Finally, these compounds exhibited neuroprotective effects, reversing partially or completely the cytotoxic effects induced by the metals studied depending on the compound used. APH4 was the most effective and safe compound.
  • Item
    Insulin Signaling Disruption and INF-γ Upregulation Induce Aβ1–42 and Hyperphosphorylated-Tau Proteins Synthesis and Cell Death after Paraquat Treatment of Primary Hippocampal Cells
    (Chemical Research in Toxicology, 2022) Abascal, Maria Luisa; Sanjuan, Javier; Moyano-Cires Ivanoff, Paula Viviana; Sola Vendrell, Emma; Flores, Andrea; García Sánchez, José Manuel; García Lobo, Jimena; Frejo Moya, María Teresa; Pino Sans, Javier Del
    Acute and long-term paraquat (PQ) exposure produces hippocampal neurodegeneration and cognition decline. Although some mechanisms involved in these effects were found, the rest are unknown. PQ treatment, for 1 and 14 days, upregulated interferon-gamma signaling, which reduced insulin levels and downregulated the insulin pathway through phosphorylated-c-Jun N-terminal-kinase upregulation, increasing glucose levels and the production of Aβ1–42 and phosphorylated-tau, by beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) overexpression and phosphorylated-GSK3β (p-GSK3β; ser9) level reduction, respectively, which induced primary hippocampal neuronal loss. This novel information on the PQ mechanisms leading to hippocampal neurodegeneration could help reveal the PQ actions that lead to cognition dysfunction.
  • Item
    Project number: 95
    Aprendizaje global, flexible y personalizado para atender la diversidad en el aula, mediante el uso de Flipped Learning, a través del uso integrado de un sistema virtual de respuesta en el aula, y otras herramientas TACs digitales
    (2021) Pino Sans, Javier Del; Moyano-Cires Ivanoff, Paula Viviana; Frejo Moya, María Teresa; Anadón Baselga, María José; Lobo Alonso, Margarita; Pelayo Alarcon, Adela; Capo Marti, Miguel Andrés; Sola Vendrell, Emma; Sanjuán López, Javier; García Sánchez, José Manuel; Ruiz Fernandez, Matilde; Mourin Moral, Francisco Javier; Frias González, Mariano De; Naval López, María Victoria; Guerra Menéndez, Lucia; García Lobo, Jimena
  • Item
    Toxicological and pharmacological evaluation, antioxidant, ADMET and molecular modeling of selected racemic chromenotacrines {11-amino-12-aryl-8,9,10,12-tetrahydro-7H-chromeno[2,3-b]quinolin-3-ols} for the potential prevention and treatment of Alzheimer’s disease
    (European Journal of Medicinal Chemistry, 2014) Oset Gasque, María Jesús; González Prieto, María Pilar; Pérez Peña, Javier; García Font, Nuria; Romero Martínez, Manuel Alejandro; Pino Sans, Javier Del; Ramos Alonso, Eva; Hadjipavlou-Litina, Dimitra; Soriano, Elena; Chioua, Mourad; Samadi, Abdelouahid; Raghuvanshi, Dushyant S.; Singh, Krishna N.; Marco Contelles, José
    The pharmacological analysis of racemic chromenotacrines (CT) 1e7, bearing the 11-amino-12-aryl-8,9,10,12-tetrahydro-7H-chromeno[2,3-b]quinolin-3-ol ring skeleton, in a series of experiments targeted to explore their potential use for the treatment of Alzheimer’s disease (AD), is reported. The toxicological evaluation showed that among all these chromenotacrines, CT6 is much less hepatotoxic than tacrine in a range of concentrations from 1 to 300 mM, measured as cell viability in HepG2 cells. Moreover, CT6 did not significantly increase lactate dehydrogenase, aspartate transaminase, and alanine transaminase release in HepG2 cells. Besides,CT6treatment exerts a high protective effect against thelipid peroxidationinduced after H2O2-treated SHSY5Y cells, in a concentration-dependent manner. CT6 showed an excellent antioxidant profile in the AAPH test, and protects against the decrease in cell viability induced by respiratory chain inhibitors (Oligomicyn A/Rotenone)and NO donors in neuronal cultures. This effect could be due to a mixed antiapoptotic and antinecrotic neuroprotective effect at low and intermediate CT6 concentrations, respectively. CT1-7 are potent and selective inhibitors of EeAChE in the submicromolar range. CT3 [IC50 (EeAChE) ¼ 0.007 0.003 mM], and CT6 [IC50 (EeAChE) ¼ 0.041 0.001 mM] are the most potent AChE inhibitors. Kinetic studies on the non-toxic chromenotacrine CT6 showed that this compound behaves as a non-competitive inhibitor (Ki ¼ 0.047 0.003 mM),indicating that CT6 binds at the peripheral anionic site, a fact confirmed by molecular modeling analysis. In silico ADMET analysis showed also that CT6 should have a moderate BBB permeability. Consequently, non-toxic chromenotacrine CT6can be considered as an ttractivemultipotent molecule for the potential treatment of AD.
  • Item
    Neurodegenerative Proteinopathies Induced by Environmental Pollutants: Heat Shock Proteins and Proteasome as Promising Therapeutic Tools
    (Pharmaceuticals, 2023) Moyano-Cires Ivanoff, Paula Viviana; Sola Vendrell, Emma; Naval López, María Victoria; Guerra Menéndez, Lucía; Fernández Fernández, María De La Cabeza; Pino Sans, Javier Del
    Environmental pollutants’ (EPs) amount and diversity have increased in recent years due to anthropogenic activity. Several neurodegenerative diseases (NDs) are theorized to be related to EPs, as their incidence has increased in a similar way to human EPs exposure and they reproduce the main ND hallmarks. EPs induce several neurotoxic effects, including accumulation and gradual deposition of misfolded toxic proteins, producing neuronal malfunction and cell death. Cells possess different mechanisms to eliminate these toxic proteins, including heat shock proteins (HSPs) and the proteasome system. The accumulation and deleterious effects of toxic proteins are induced through HSPs and disruption of proteasome proteins’ homeostatic function by exposure to EPs. A therapeutic approach has been proposed to reduce accumulation of toxic proteins through treatment with recombinant HSPs/proteasome or the use of compounds that increase their expression or activity. Our aim is to review the current literature on NDs related to EP exposure and their relationship with the disruption of the proteasome system and HSPs, as well as to discuss the toxic effects of dysfunction of HSPs and proteasome and the contradictory effects described in the literature. Lastly, we cover the therapeutic use of developed drugs and recombinant proteasome/HSPs to eliminate toxic proteins and prevent/treat EP-induced neurodegeneration.
  • Item
    Manganese induced ROS and AChE variants alteration leads to SN56 basal forebrain cholinergic neuronal loss after acute and long-term treatment
    (Food and Chemical Toxicology, 2019) Moyano-Cires Ivanoff, Paula Viviana; García Sánchez, José Manuel; Anadón Baselga, María José; Lobo Alonso, Margarita; García Lobo, Jimena; Frejo Moya, María Teresa; Sola Vendrell, Emma; Pelayo Alarcón, Adela; Pino Sans, Javier Del
    Manganese (Mn) induces cognitive disorders and basal forebrain (BF) cholinergic neuronal loss, involved on learning and memory regulation, which could be the cause of such cognitive disorders. However, the mechanisms through which it induces these effects are unknown. We hypothesized that Mn could induce BF cholinergic neuronal loss through oxidative stress generation, cholinergic transmission and AChE variants alteration that could explain Mn cognitive disorders. This study shows that Mn impaired cholinergic transmission in SN56 cholinergic neurons from BF through alteration of AChE and ChAT activity and CHT expression. Moreover, Mn induces, after acute and long-term exposure, AChE variants alteration and oxidative stress generation that leaded to lipid peroxidation and protein oxidation. Finally, Mn induces cell death on SN56 cholinergic neurons and this effect is independent of cholinergic transmission alteration, but was mediated partially by oxidative stress generation and AChE variants alteration. Our results provide new understanding of the mechanisms contributing to the harmful effects of Mn on cholinergic neurons and their possible involvement in cognitive disorders induced by Mn.