Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Arch Toxicol ; 91(1): 407-425, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26928308

RESUMEN

Halogen-free organophosphorus flame retardants are considered as replacements for the phased-out class of polybrominated diphenyl ethers (PBDEs). However, toxicological information on new flame retardants is still limited. Based on their excellent flame retardation potential, we have selected three novel 9,10-dihydro-9-oxa-10-phosphaphenanthrene-10-oxide (DOPO) derivatives and assessed their toxicological profile using a battery of in vitro test systems in order to provide toxicological information before their large-scale production and use. PBDE-99, applied as a reference compound, exhibited distinct neuro-selective cytotoxicity at concentrations ≥10 µM. 6-(2-((6-oxido-6H-dibenzo[c,e][1,2]oxaphosphinin-6-yl)amino)ethoxy)-6H-dibenzo[c,e][1,2]oxaphosphinine 6-oxide (ETA-DOPO) and 6,6'-(ethane-1,2-diylbis(oxy))bis(6H-dibenzo[c,e][1,2]oxaphosphinine-6-oxide) (EG-DOPO) displayed adverse effects at concentrations >10 µM in test systems reflecting the properties of human central and peripheral nervous system neurons, as well as in a set of non-neuronal cell types. DOPO and its derivative 6,6'-(ethane-1,2-diylbis(azanediyl))bis(6H-dibenzo[c,e][1,2]oxaphosphinine-6-oxide) (EDA-DOPO) were neither neurotoxic, nor did they exhibit an influence on neural crest cell migration, or on the integrity of human skin equivalents. The two compounds furthermore displayed no inflammatory activation potential, nor did they affect algae growth or daphnia viability at concentrations ≤400 µM. Based on the superior flame retardation properties, biophysical features suited for use in polyurethane foams, and low cytotoxicity of EDA-DOPO, our results suggest that it is a candidate for the replacement of currently applied flame retardants.


Asunto(s)
Retardadores de Llama/toxicidad , Queratinocitos/efectos de los fármacos , Monocitos/efectos de los fármacos , Neuronas/efectos de los fármacos , Compuestos Organofosforados/toxicidad , Mucosa Respiratoria/efectos de los fármacos , Piel/efectos de los fármacos , Células A549 , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Óxidos P-Cíclicos/toxicidad , Células Madre Embrionarias Humanas/citología , Humanos , Queratinocitos/citología , Queratinocitos/inmunología , Queratinocitos/metabolismo , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Neuronas/citología , Neuronas/inmunología , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Piel/citología , Piel/inmunología , Piel/metabolismo , Absorción Cutánea , Pruebas de Irritación de la Piel , Sus scrofa , Andamios del Tejido/química , Pruebas de Toxicidad
2.
Redox Biol ; 73: 103165, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38688061

RESUMEN

The activation of ferroptosis is being pursued in cancer research as a strategy to target apoptosis-resistant cells. By contrast, in various diseases that affect the cardiovascular system, kidneys, liver, and central and peripheral nervous systems, attention is directed toward interventions that prevent ferroptotic cell death. Mechanistic insights into both research areas stem largely from studies using cellular in vitro models. However, intervention strategies that show promise in cellular test systems often fail in clinical trials, which raises concerns regarding the predictive validity of the utilized in vitro models. In this study, the human LUHMES cell line, which serves as a model for human dopaminergic neurons, was used to characterize factors influencing the activation of ferroptosis. Erastin and RSL-3 induced cell death that was distinct from apoptosis. Parameters such as the differentiation state of LUHMES cells, cell density, and the number and timing of medium changes were identified as determinants of sensitivity to ferroptosis activation. In differentiated LUHMES cells, interventions at mechanistically divergent sites (iron chelation, coenzyme Q10, peroxidase mimics, or inhibition of 12/15-lipoxygenase) provide almost complete protection from ferroptosis. LUHMES cells allowed the experimental modulation of intracellular iron concentrations and demonstrated a correlation between intracellular iron levels, the rate of lipid peroxidation, as well as the sensitivity of the cells to ferroptotic cell death. These findings underscore the importance of understanding the various factors that influence ferroptosis activation and highlight the need for well-characterized in vitro models to enhance the reliability and predictive value of observations in ferroptosis research, particularly when translating findings into in vivo contexts.


Asunto(s)
Neuronas Dopaminérgicas , Ferroptosis , Humanos , Neuronas Dopaminérgicas/metabolismo , Línea Celular , Piperazinas/farmacología , Hierro/metabolismo , Diferenciación Celular , Apoptosis , Carbolinas , Ubiquinona/análogos & derivados
3.
J Biol Chem ; 286(7): 4991-5002, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21081502

RESUMEN

Minocycline prevents oxidative protein modifications and damage in disease models associated with inflammatory glial activation and oxidative stress. Although the drug has been assumed to act by preventing the up-regulation of proinflammatory enzymes, we probed here its direct chemical interaction with reactive oxygen species. The antibiotic did not react with superoxide or (•)NO radicals, but peroxynitrite (PON) was scavenged in the range of ∼1 µm minocycline and below. The interaction of pharmacologically relevant minocycline concentrations with PON was corroborated in several assay systems and significantly exceeded the efficacy of other antibiotics. Minocycline was degraded during the reaction with PON, and the resultant products lacked antioxidant properties. The antioxidant activity of minocycline extended to cellular systems, because it prevented neuronal mitochondrial DNA damage and glutathione depletion. Maintenance of neuronal viability under PON stress was shown to be solely dependent on direct chemical scavenging by minocycline. We chose α-synuclein (ASYN), known from Parkinsonian pathology as a biologically relevant target in chemical and cellular nitration reactions. Submicromolar concentrations of minocycline prevented tyrosine nitration of ASYN by PON. Mass spectrometric analysis revealed that minocycline impeded nitrations more effectively than methionine oxidations and dimerizations of ASYN, which are secondary reactions under PON stress. Thus, PON scavenging at low concentrations is a novel feature of minocycline and may help to explain its pharmacological activity.


Asunto(s)
Minociclina/química , Fármacos Neuroprotectores/química , Ácido Peroxinitroso/química , Antibacterianos/química , Antibacterianos/farmacología , Línea Celular Transformada , Daño del ADN/efectos de los fármacos , ADN Mitocondrial/química , ADN Mitocondrial/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Minociclina/farmacología , Neuronas/química , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Ácido Peroxinitroso/metabolismo , Multimerización de Proteína/efectos de los fármacos , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo
4.
Trends Pharmacol Sci ; 38(6): 541-555, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28442167

RESUMEN

The neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) causes a Parkinson's disease (PD)-like syndrome by inducing degeneration of nigrostriatal dopaminergic neurons. Studies of the MPTP model have revealed the pathomechanisms underlying dopaminergic neurodegeneration and facilitated the development of drug treatments for PD. In this review, we provide an update on MPTP bioactivation and biodistribution, reconcile the distinct views on energetic failure versus reactive oxygen species (ROS) formation as main drivers of MPTP-induced neurodegeneration, and describe recently identified intrinsic features of the nigrostriatal system that make it particularly vulnerable to MPTP. We discuss these new perspectives on the endogenous tipping points of tissue homeostasis and the drivers responsible for vicious cycles in relation to their relevance for the development of novel intervention strategies for PD.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Intoxicación por MPTP/inducido químicamente , Enfermedades Neurodegenerativas/inducido químicamente , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacocinética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Neostriado/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología
5.
Br J Pharmacol ; 172(16): 4119-32, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25989025

RESUMEN

BACKGROUND AND PURPOSE: Few neuropharmacological model systems use human neurons. Moreover, available test systems rarely reflect functional roles of co-cultured glial cells. There is no human in vitro counterpart of the widely used 1-methyl-4-phenyl-tetrahydropyridine (MPTP) mouse model of Parkinson's disease EXPERIMENTAL APPROACH: We generated such a model by growing an intricate network of human dopaminergic neurons on a dense layer of astrocytes. In these co-cultures, MPTP was metabolized to 1-methyl-4-phenyl-pyridinium (MPP(+) ) by the glial cells, and the toxic metabolite was taken up through the dopamine transporter into neurons. Cell viability was measured biochemically and by quantitative neurite imaging, siRNA techniques were also used. KEY RESULTS: We initially characterized the activation of PARP. As in mouse models, MPTP exposure induced (poly-ADP-ribose) synthesis and neurodegeneration was blocked by PARP inhibitors. Several different putative neuroprotectants were then compared in mono-cultures and co-cultures. Rho kinase inhibitors worked in both models; CEP1347, ascorbic acid or a caspase inhibitor protected mono-cultures from MPP(+) toxicity, but did not protect co-cultures, when used alone or in combination. Application of GSSG prevented degeneration in co-cultures, but not in mono-cultures. The surprisingly different pharmacological profiles of the models suggest that the presence of glial cells, and the in situ generation of the toxic metabolite MPP(+) within the layered cultures played an important role in neuroprotection. CONCLUSIONS AND IMPLICATIONS: Our new model system is a closer model of human brain tissue than conventional cultures. Its use for screening of candidate neuroprotectants may increase the predictiveness of a test battery.


Asunto(s)
1-Metil-4-fenilpiridinio/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Intoxicación por MPTP/prevención & control , Neuroglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Neurotoxinas/toxicidad , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Neuronas Dopaminérgicas/metabolismo , Humanos , Isoquinolinas/farmacología , Intoxicación por MPTP/metabolismo , Ratones , Neuroglía/metabolismo , Neurotoxinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Ratas
6.
Antioxid Redox Signal ; 23(13): 1001-16, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26413876

RESUMEN

AIMS: 1-Methyl-4-phenyl-tetrahydropyridine (MPTP) is among the most widely used neurotoxins for inducing experimental parkinsonism. MPTP causes parkinsonian symptoms in mice, primates, and humans by killing a subpopulation of dopaminergic neurons. Extrapolations of data obtained using MPTP-based parkinsonism models to human disease are common; however, the precise mechanism by which MPTP is converted into its active neurotoxic metabolite, 1-methyl-4-phenyl-pyridinium (MPP(+)), has not been fully elucidated. In this study, we aimed to address two unanswered questions related to MPTP toxicology: (1) Why are MPTP-converting astrocytes largely spared from toxicity? (2) How does MPP(+) reach the extracellular space? RESULTS: In MPTP-treated astrocytes, we discovered that the membrane-impermeable MPP(+), which is generally assumed to be formed inside astrocytes, is almost exclusively detected outside of these cells. Instead of a transporter-mediated export, we found that the intermediate, 1-methyl-4-phenyl-2,3-dihydropyridinium (MPDP(+)), and/or its uncharged conjugate base passively diffused across cell membranes and that MPP(+) was formed predominately by the extracellular oxidation of MPDP(+) into MPP(+). This nonenzymatic extracellular conversion of MPDP(+) was promoted by O2, a more alkaline pH, and dopamine autoxidation products. INNOVATION AND CONCLUSION: Our data indicate that MPTP metabolism is compartmentalized between intracellular and extracellular environments, explain the absence of toxicity in MPTP-converting astrocytes, and provide a rationale for the preferential formation of MPP(+) in the extracellular space. The mechanism of transporter-independent extracellular MPP(+) formation described here indicates that extracellular genesis of MPP(+) from MPDP is a necessary prerequisite for the selective uptake of this toxin by catecholaminergic neurons.


Asunto(s)
1-Metil-4-fenilpiridinio/metabolismo , Neuronas Dopaminérgicas/metabolismo , Enfermedad de Parkinson/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/metabolismo , Astrocitos/metabolismo , Transporte Biológico , Catecolaminas/metabolismo , Línea Celular , Membrana Celular/metabolismo , Difusión , Líquido Extracelular/metabolismo , Humanos , Monoaminooxidasa/metabolismo , Oxidación-Reducción , Enfermedad de Parkinson/patología , Compuestos de Piridinio/metabolismo
7.
ALTEX ; 29(3): 261-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22847254

RESUMEN

Astrocytes are activated in most chronic neurodegenerative diseases associated with inflammatory events such as Parkinson's disease or Alzheimer's disease, but also in stroke. Due to an aging population worldwide, research efforts in these areas are likely to expand in the future. This will entail an increased demand for appropriate experimental models. We introduce here the new immortalized mouse astrocyte cell line IMA 2.1 as an alternative to currently used primary astrocyte cultures. IMA 2.1 were directly compared with primary mouse astrocytes with respect to their response to proinflammatory stimuli, expression of typical astrocyte markers, and to the cell line's capacity to metabolize the parkinsonian toxin MPTP to its toxic metabolite MPP+. Under inflammatory conditions, mimicked with the addition of a cytokine mix, IMA 2.1 responded similarly to primary astrocytes with mRNA upregulation, expression of iNOS and COX-2, and the release of various inflammatory mediators. Analysis of astrocytic markers indicated that IMA 2.1 represent a relatively early, GFAP-negative stage of astrocyte development. Moreover, conversion of MPTP by monoamine oxidase-B proceeded in IMA at least as quickly as in primary cells. For all endpoints investigated, the cell line IMA 2.1, derived from a single clone, delivered reproducible results over a period of several years and allowed upscaling of experiments due to its easy handling compared with primary cells.


Asunto(s)
Astrocitos/citología , Astrocitos/fisiología , Animales , Línea Celular , Técnicas de Cocultivo , Citometría de Flujo , Regulación de la Expresión Génica/fisiología , Humanos , Inflamación , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA