Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Int J Mol Sci ; 24(5)2023 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-36901772

RESUMEN

Exposure to chemicals may pose a greater risk to vulnerable groups, including pregnant women, fetuses, and children, that may lead to diseases linked to the toxicants' target organs. Among chemical contaminants, methylmercury (MeHg), present in aquatic food, is one of the most harmful to the developing nervous system depending on time and level of exposure. Moreover, certain man-made PFAS, such as PFOS and PFOA, used in commercial and industrial products including liquid repellants for paper, packaging, textile, leather, and carpets, are developmental neurotoxicants. There is vast knowledge about the detrimental neurotoxic effects induced by high levels of exposure to these chemicals. Less is known about the consequences that low-level exposures may have on neurodevelopment, although an increasing number of studies link neurotoxic chemical exposures to neurodevelopmental disorders. Still, the mechanisms of toxicity are not identified. Here we review in vitro mechanistic studies using neural stem cells (NSCs) from rodents and humans to dissect the cellular and molecular processes changed by exposure to environmentally relevant levels of MeHg or PFOS/PFOA. All studies show that even low concentrations dysregulate critical neurodevelopmental steps supporting the idea that neurotoxic chemicals may play a role in the onset of neurodevelopmental disorders.


Asunto(s)
Compuestos de Metilmercurio , Células-Madre Neurales , Síndromes de Neurotoxicidad , Niño , Humanos , Femenino , Embarazo , Compuestos de Metilmercurio/toxicidad , Sustancias Peligrosas/farmacología
2.
Environ Toxicol ; 37(8): 2044-2057, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35485992

RESUMEN

Glyphosate-based herbicides (GBH) are among the most sold pesticides in the world. There are several formulations based on the active ingredient glyphosate (GLY) used along with other chemicals to improve the absorption and penetration in plants. The final composition of commercial GBH may modify GLY toxicological profile, potentially enhancing its neurotoxic properties. The developing nervous system is particularly susceptible to insults occurring during the early phases of development, and exposure to chemicals in this period may lead to persistent impairments on neurogenesis and differentiation. The aim of this study was to evaluate the long-lasting effects of a sub-cytotoxic concentration, 2.5 parts per million of GBH and GLY, on the differentiation of human neuroepithelial stem cells (NES) derived from induced pluripotent stem cells (iPSC). We treated NES cells with each compound and evaluated the effects on key cellular processes, such as proliferation and differentiation in daughter cells never directly exposed to the toxicants. We found that GBH induced a more immature neuronal profile associated to increased PAX6, NESTIN and DCX expression, and a shift in the differentiation process toward glial cell fate at the expense of mature neurons, as shown by an increase in the glial markers GFAP, GLT1, GLAST and a decrease in MAP2. Such alterations were associated to dysregulation of key genes critically involved in neurogenesis, including PAX6, HES1, HES5, and DDK1. Altogether, the data indicate that subtoxic concentrations of GBH, but not of GLY, induce long-lasting impairments on the differentiation potential of NES cells.


Asunto(s)
Herbicidas , Glicina/análogos & derivados , Glicina/toxicidad , Herbicidas/toxicidad , Humanos , Neurogénesis , Neuronas , Glifosato
3.
Biochem Biophys Res Commun ; 482(3): 408-418, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28212724

RESUMEN

Epidemiological and/or experimental studies have shown that unfavorable prenatal environmental factors, such as stress or exposure to certain neurotoxic environmental contaminants, may have adverse consequences for neurodevelopment. Alterations in neurogenesis can have harmful effects not only for the developing nervous system, but also for the adult brain where neurogenesis is believed to play a role in learning, memory, and even in depression. Many recent advances in the understanding of the complex process of nervous system development can be integrated into the field of neurotoxicology. In the past 15 years we have been using cultured neural stem or progenitor cells to investigate the effects of neurotoxic stimuli on cell survival, proliferation and differentiation, with special focus on heritable effects. This is an overview of the work performed by our group in the attempt to elucidate the mechanisms of developmental neurotoxicity and possibly provide relevant information for the understanding of the etiopathogenesis of complex brain disorders.


Asunto(s)
Síndromes de Neurotoxicidad/etiología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Humanos , Modelos Neurológicos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Neurogénesis/efectos de los fármacos , Neurotoxinas/toxicidad
4.
Toxicol Appl Pharmacol ; 269(1): 51-60, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23500012

RESUMEN

Perfluorinated compounds are ubiquitous chemicals of major concern for their potential adverse effects on the human population. We have used primary rat embryonic neural stem cells (NSCs) to study the effects of perfluorooctane sulfonate (PFOS) on the process of NSC spontaneous differentiation. Upon removal of basic fibroblast growth factor, NSCs were exposed to nanomolar concentrations of PFOS for 48 h, and then allowed to differentiate for additional 5 days. Exposure to 25 or 50 nM concentration resulted in a lower number of proliferating cells and a higher number of neurite-bearing TuJ1-positive cells, indicating an increase in neuronal differentiation. Exposure to 50 nM also significantly increased the number of CNPase-positive cells, pointing to facilitation of oligodendrocytic differentiation. PPAR genes have been shown to be involved in PFOS toxicity. By q-PCR we detected an upregulation of PPARγ with no changes in PPARα or PPARδ genes. One of the downstream targets of PPARs, the mitochondrial uncoupling protein 2 (UCP2) was also upregulated. The number of TuJ1- and CNPase-positive cells increased after exposure to PPARγ agonist rosiglitazone (RGZ, 3 µM) and decreased after pre-incubation with the PPARγ antagonist GW9662 (5 µM). RGZ also upregulated the expression of PPARγ and UCP2 genes. Meanwhile GW9662 abolished the UCP2 upregulation and decreased Ca²âº activity induced by PFOS. Interestingly, a significantly higher expression of PPARγ and UCP3 genes was also detected in mouse neonatal brain after prenatal exposure to PFOS. These data suggest that PPARγ plays a role in the alteration of spontaneous differentiation of NSCs induced by nanomolar concentrations of PFOS.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Encéfalo/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , PPAR gamma/efectos de los fármacos , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Regulación de la Expresión Génica/efectos de los fármacos , Canales Iónicos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/metabolismo , Oligodendroglía/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Proteína Desacopladora 2 , Proteína Desacopladora 3
5.
Mol Neurobiol ; 60(12): 7346-7361, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37561236

RESUMEN

Exposure to prenatal insults, such as excess glucocorticoids (GC), may lead to pathological outcomes, including neuropsychiatric disorders. The aim of the present study was to investigate the long-term effects of in utero exposure to the synthetic GC analog dexamethasone (Dex) in adult female offspring. We monitored spontaneous activity in the home cage under a constant 12 h/12 h light/dark cycle, as well as the changes following a 6-h advance of dark onset (phase shift). For comparison, we re-analysed data previously recorded in males. Dex-exposed females were spontaneously more active, and the activity onset re-entrained slower than in controls. In contrast, Dex-exposed males were less active, and the activity onset re-entrained faster than in controls. Following the phase shift, control females displayed a transient reorganisation of behaviour in light and virtually no change in dark, while Dex-exposed females showed limited variations from baseline in both light and dark, suggesting weaker photic entrainment. Next, we ran bulk RNA-sequencing in the suprachiasmatic nucleus (SCN) of Dex and control females. SPIA pathway analysis of ~ 2300 differentially expressed genes identified significantly downregulated dopamine signalling, and upregulated glutamate and GABA signalling. We selected a set of candidate genes matching the behaviour alterations and found consistent differential regulation for ~ 73% of tested genes in SCN and hippocampus tissue samples. Taken together, our data highlight sex differences in the outcome of prenatal exposure to excess GC in adult mice: in contrast to depression-like behaviour in males, the phenotype in females, defined by behaviour and differential gene expression, is consistent with ADHD models.


Asunto(s)
Trastornos Mentales , Efectos Tardíos de la Exposición Prenatal , Humanos , Embarazo , Femenino , Ratones , Masculino , Animales , Glucocorticoides/farmacología , Caracteres Sexuales , Efectos Tardíos de la Exposición Prenatal/metabolismo , Núcleo Supraquiasmático/metabolismo , Trastornos Mentales/metabolismo , Ritmo Circadiano/fisiología
6.
Neurotoxicology ; 83: 1-13, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33338551

RESUMEN

Clinical and preclinical evidence indicates that prenatal exposure to glucocorticoids may induce detrimental effects in the offspring, including reduction in fetal growth and alterations in the CNS. On this basis, the present study investigated whether in utero exposure to high levels of glucocorticoids is a risk factor that may lead to an exacerbation of the central noxious effects induced by psychoactive drugs consumed later in life. To this end, pregnant C57BL6/J dams were treated with dexamethasone (DEX, 0.05 mg/kg per day) from gestational day 14 until delivery. Thereafter, the male offspring were evaluated to ascertain the magnitude of dopaminergic damage, astrogliosis and microgliosis elicited in the nigrostriatal tract by the amphetamine-related drug 3,4--methylenedioxymethamphetamine (MDMA, 4 × 20 mg/kg, 2 h apart, sacrificed 48 h later) administered at either adolescence or adulthood. Immunohistochemistry was performed in the substantia nigra pars compacta (SNc) and striatum, to evaluate dopaminergic degeneration by measuring tyrosine hydroxylase (TH), as well as astrogliosis and microgliosis by measuring glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA-1), respectively. Moreover, immunohistochemistry was used to ascertain the co-localization of IBA-1 with either the pro-inflammatory interleukin (IL) IL-1ß or the anti-inflammatory IL IL-10, in order to determine the microglial phenotype. In utero administration of DEX induced dopaminergic damage by decreasing the density of TH-positive fibers in the striatum, although only in adult mice. MDMA administration induced dopaminergic damage and glia activation in the nigrostriatal tract of adolescent and adult mice. Mice exposed to DEX in utero and treated with MDMA later in life showed a more pronounced loss of dopaminergic neurons (adolescent mice) and astrogliosis (adolescent and adult mice) in the SNc, compared with control mice. These results suggest that prenatal exposure to glucocorticoids may induce an age-dependent and persistent increase in the susceptibility to central toxicity of amphetamine-related drugs used later in life.


Asunto(s)
Encéfalo/efectos de los fármacos , Dexametasona/toxicidad , Neuronas Dopaminérgicas/efectos de los fármacos , Glucocorticoides/toxicidad , Neuroglía/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Femenino , Edad Gestacional , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Masculino , Exposición Materna , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , N-Metil-3,4-metilenodioxianfetamina , Degeneración Nerviosa , Neuroglía/metabolismo , Neuroglía/patología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Embarazo , Tirosina 3-Monooxigenasa/metabolismo
7.
EFSA J ; 18(9): e06223, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32994824

RESUMEN

The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluoroalkyl substances (PFASs) in food. Based on several similar effects in animals, toxicokinetics and observed concentrations in human blood, the CONTAM Panel decided to perform the assessment for the sum of four PFASs: PFOA, PFNA, PFHxS and PFOS. These made up half of the lower bound (LB) exposure to those PFASs with available occurrence data, the remaining contribution being primarily from PFASs with short half-lives. Equal potencies were assumed for the four PFASs included in the assessment. The mean LB exposure in adolescents and adult age groups ranged from 3 to 22, the 95th percentile from 9 to 70 ng/kg body weight (bw) per week. Toddlers and 'other children' showed a twofold higher exposure. Upper bound exposure was 4- to 49-fold higher than LB levels, but the latter were considered more reliable. 'Fish meat', 'Fruit and fruit products' and 'Eggs and egg products' contributed most to the exposure. Based on available studies in animals and humans, effects on the immune system were considered the most critical for the risk assessment. From a human study, a lowest BMDL 10 of 17.5 ng/mL for the sum of the four PFASs in serum was identified for 1-year-old children. Using PBPK modelling, this serum level of 17.5 ng/mL in children was estimated to correspond to long-term maternal exposure of 0.63 ng/kg bw per day. Since accumulation over time is important, a tolerable weekly intake (TWI) of 4.4 ng/kg bw per week was established. This TWI also protects against other potential adverse effects observed in humans. Based on the estimated LB exposure, but also reported serum levels, the CONTAM Panel concluded that parts of the European population exceed this TWI, which is of concern.

8.
Neurosignals ; 17(3): 203-12, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19346757

RESUMEN

Akt has been implicated in pro-survival and anti-apoptotic activities in many cell types, including dorsal root ganglion (DRG) and spinal motor neurons. In this immunohistochemical study we have monitored phosphorylated Akt (p-Akt) levels in adult mouse DRGs and spinal cord following unilateral peripheral sciatic nerve transection (axotomy) or carrageenan-induced inflammation. In control animals around half of the lumbar DRG neuron profiles (NPs), mainly small and medium-sized ones, were p-Akt immunoreactive (IR), and of these around 50% expressed calcitonin gene-related peptide and/or isolectin IB4. Two weeks after axotomy, the number of p-Akt-positive NPs was only slightly reduced, but p-Akt immunofluorescence intensity was strongly increased. One third of the ipsilateral p-Akt-IR NPs was galanin positive, but virtually without colocalization with neuropeptide Y. Furthermore, p-Akt-like immunoreactivity significantly increased in intensity in the ipsilateral spinal dorsal horn after axotomy and expanded into deeper layers. Carrageenan-induced peripheral inflammation increased the number of p-Akt-IR NPs after 1 h. Both axotomy and inflammation caused a clear increase in nuclear p-Akt-like immunoreactivity in DRG neurons. Our findings support a role for Akt as a key signaling molecule in sensory neurons and spinal cord after peripheral injury.


Asunto(s)
Ganglios Espinales/metabolismo , Neuralgia/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Receptoras Sensoriales/metabolismo , Médula Espinal/metabolismo , Animales , Axotomía , Péptido Relacionado con Gen de Calcitonina/metabolismo , Núcleo Celular/metabolismo , Tamaño de la Célula , Galanina/metabolismo , Ganglios Espinales/citología , Inmunohistoquímica , Mediadores de Inflamación/farmacología , Vértebras Lumbares , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/patología , Neuralgia/fisiopatología , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Fosforilación , Nervio Ciático/metabolismo , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología , Neuropatía Ciática/fisiopatología , Células Receptoras Sensoriales/citología , Transducción de Señal/fisiología , Médula Espinal/citología
9.
Transl Psychiatry ; 9(1): 263, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31624238

RESUMEN

Alterations in circadian rhythms are closely linked to depression, and we have shown earlier that progressive alterations in circadian entrainment precede the onset of depression in mice exposed in utero to excess glucocorticoids. The aim of this study was to investigate whether treatment with the noradrenaline reuptake inhibitor desipramine (DMI) could restore the alterations in circadian entrainment and prevent the onset of depression-like behavior. C57Bl/6 mice were exposed to dexamethasone (DEX-synthetic glucocorticoid analog, 0.05 mg/kg/day) between gestational day 14 and delivery. Male offspring aged 6 months (mo) were treated with DMI (10 mg/kg/day in drinking water) for at least 21 days before behavioral testing. We recorded spontaneous activity using the TraffiCage™ system and found that DEX mice re-entrained faster than controls after an abrupt advance in light-dark cycle by 6 h, while DMI treatment significantly delayed re-entrainment. Next we assessed the synchronization of peripheral oscillators with the central clock (located in the suprachiasmatic nucleus-SCN), as well as the mechanisms required for entrainment. We found that photic entrainment of the SCN was apparently preserved in DEX mice, but the expression of clock genes in the hippocampus was not synchronized with the light-dark cycle. This was associated with downregulated mRNA expression for arginine vasopressin (AVP; the main molecular output entraining peripheral clocks) in the SCN, and for glucocorticoid receptor (GR; required for the negative feedback loop regulating glucocorticoid secretion) in the hippocampus. DMI treatment restored the mRNA expression of AVP in the SCN and enhanced GR-mediated signaling by upregulating GR expression and nuclear translocation in the hippocampus. Furthermore, DMI treatment at 6 mo prevented the onset of depression-like behavior and the associated alterations in neurogenesis in 12-mo-old DEX mice. Taken together, our data indicate that DMI treatment enhances GR-mediated signaling and restores the synchronization of peripheral clocks with the SCN and support the hypothesis that altered circadian entrainment is a modifiable risk factor for depression.


Asunto(s)
Antidepresivos Tricíclicos/administración & dosificación , Ritmo Circadiano/efectos de los fármacos , Depresión/prevención & control , Desipramina/administración & dosificación , Dexametasona/toxicidad , Glucocorticoides/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Depresión/inducido químicamente , Femenino , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Neurogénesis/efectos de los fármacos , Fotoperiodo , Embarazo , Núcleo Supraquiasmático/efectos de los fármacos
10.
J Neurosci Methods ; 311: 259-266, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389486

RESUMEN

BACKGROUND: The spinal cord is composed of a large number of cells that interact to allow the organism to function. To perform detail studies of cellular processes involved in spinal cord injury (SCI), one must use repeatable and specific methods to target and injure restricted areas of the spinal cord. NEW METHOD: We propose a robust method to induce SCI in zebrafish by laser light. With a 2-photon microscope equipped with a femtosecond near-infrared pump laser, we explored the effects of laser beam exposure time, area, and intensity to induce precise and repeatable SCI with minimized collateral damage to neighboring cells. RESULTS: Through behavioral studies in zebrafish larvae, we assessed the functional outcome of intensive laser light directed at the spinal cord. Our experiments revealed that a laser pulse with wavelength 800 nm, duration 2.6 ms, and light intensity 390 mW was sufficient to induce controlled cell death in a single cell or a spinal cord segment. Collateral damage was observed if cells were exposed to laser pulses exceeding 470 mW. With these settings, we could induce precise and repeatable SCI in zebrafish larvae, resulting in loss of motor and sensory function. COMPARISON WITH EXISTING METHOD(S): Our method offers a simple and more controlled setting to induce SCI in zebrafish. We describe how the near-infrared femtosecond laser should be adjusted for achieving optimal results with minimal collateral damage. CONCLUSIONS: We present a precise and robust method for inducing SCI in zebrafish with single-cell resolution using femtosecond near-infrared laser pulses.


Asunto(s)
Modelos Animales de Enfermedad , Rayos Láser , Procedimientos Neuroquirúrgicos/instrumentación , Procedimientos Neuroquirúrgicos/métodos , Traumatismos de la Médula Espinal/fisiopatología , Animales , Larva , Locomoción , Microscopía Confocal/métodos , Microcirugia/instrumentación , Microcirugia/métodos , Traumatismos de la Médula Espinal/patología , Pez Cebra
12.
Front Genet ; 10: 593, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31316548

RESUMEN

Controversial evidence points to a possible involvement of methylmercury (MeHg) in the etiopathogenesis of autism spectrum disorders (ASD). In the present study, we used human neuroepithelial stem cells from healthy donors and from an autistic patient bearing a bi-allelic deletion in the gene encoding for NRXN1 to evaluate whether MeHg would induce cellular changes comparable to those seen in cells derived from the ASD patient. In healthy cells, a subcytotoxic concentration of MeHg enhanced astroglial differentiation similarly to what observed in the diseased cells (N1), as shown by the number of GFAP positive cells and immunofluorescence signal intensity. In both healthy MeHg-treated and N1 untreated cells, aberrations in Notch pathway activity seemed to play a critical role in promoting the differentiation toward glia. Accordingly, treatment with the established Notch inhibitor DAPT reversed the altered differentiation. Although our data are not conclusive since only one of the genes involved in ASD is considered, the results provide novel evidence suggesting that developmental exposure to MeHg, even at subcytotoxic concentrations, induces alterations in astroglial differentiation similar to those observed in ASD.

13.
J Neurochem ; 106(3): 1378-87, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18485098

RESUMEN

Substantial evidence indicates that predisposition to diseases can be acquired during early stages of development and interactions between environmental and genetic factors may be implicated in the onset of many pathological conditions. Data collected over several decades have shown that chemicals are among the relevant factors that can endanger CNS. We previously showed that perinatal exposure to methylmercury (MeHg) causes persistent changes in learning and motivational behavior in mice. In this study, we report that the depression-like behavior in MeHg-exposed male mice is reversed by chronic treatment with the antidepressant fluoxetine. Behavioral alterations are associated with a decrease in brain-derived neurotrophic factor (BDNF) mRNA in the hippocampal dentate gyrus and fluoxetine treatment restores BDNF mRNA expression. We also show that MeHg-exposure induces long-lasting repressive state of the chromatin structure at the BDNF promoter region, in particular DNA hypermethylation, an increase in histone H3-K27 tri-methylation and a decrease in H3 acetylation at the promoter IV. While fluoxetine treatment does not alter hypermethylation of H3-K27, it significantly up-regulates H3 acetylation at the BDNF promoter IV in MeHg-exposed mice. Our study shows that developmental exposure to low levels of MeHg predisposes mice to depression and induces epigenetic suppression of BDNF gene expression in the hippocampus.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Depresión/metabolismo , Epigénesis Genética/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Compuestos de Metilmercurio/toxicidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Depresión/inducido químicamente , Depresión/tratamiento farmacológico , Epigénesis Genética/fisiología , Femenino , Fluoxetina/uso terapéutico , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmovilización , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética
14.
Apoptosis ; 13(3): 354-63, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18181021

RESUMEN

Oxidative stress occurs as a consequence of disturbance in the balance between the generation of reactive oxygen species (ROS) and the antioxidant defence mechanisms. The interaction of ROS with DNA can cause single-, or double-strand breaks that subsequently can lead to the activation of p53, which is central for the regulation of cellular response, e.g. apoptosis, to a range of environmental and intracellular stresses. Previous reports have suggested a regulatory role of p53 in the early activation of caspase-2, upstream of mitochondrial apoptotic signaling. Here we show that excessive ROS formation, induced by 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) exposure, induces apoptosis in primary cultured neural stem cells (NSCs) from cortices of E15 rat embryos. Following DMNQ exposure cells exhibited apoptotic hallmarks such as Bax oligomerization and activation, cytochrome c release, caspase activation and chromatin condensation. Additionally, we could show early p53 accumulation and a subsequent activation of caspase-2. The attenuation of caspase-2 activity with selective inhibitors could antagonize the mitochondrial signaling pathway and cell death. Overall, our results strongly suggest that DMNQ-induced oxidative stress causes p53 accumulation and consequently caspase-2 activation, which in turn initiates apoptotic cell death via the mitochondria-mediated caspase-dependent pathway in NSCs.


Asunto(s)
Apoptosis/fisiología , Cisteína Endopeptidasas/metabolismo , Células Madre Embrionarias/enzimología , Estrés Oxidativo/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Madre Embrionarias/efectos de los fármacos , Activación Enzimática , Naftoquinonas/toxicidad , Neuronas/citología , Estrés Oxidativo/efectos de los fármacos , Ratas , Regulación hacia Arriba
15.
Neuroreport ; 19(3): 339-43, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18303578

RESUMEN

The Notch receptor is essential for neural stem cell (NSC) characteristics. Relatively high concentrations (micromolar) of methylmercury (MeHg) activate Notch signalling in Drosophila cell lines; however, exposure of MeHg at such concentrations is rare, and the implications for mammalian cells are unclear. We have shown that MeHg at a nanomolar range inhibits neuronal differentiation of rodent embryonic NSCs. Here we show that low MeHg levels (2.5-10 nM) activated Notch signalling in NSCs, as assessed by the increased activity in a specific Notch-reporter assay and by the increased cleavage of the Notch intracellular domain. Importantly, pretreatment with Notch cleavage inhibitor reversed the MeHg-induced repression of neuronal differentiation, suggesting that Notch activation is involved in the inhibition of NSC differentiation by environmentally relevant levels of MeHg.


Asunto(s)
Compuestos de Metilmercurio/toxicidad , Neuronas/efectos de los fármacos , Receptores Notch/fisiología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Dipéptidos/farmacología , Femenino , Manganeso/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Embarazo , Inhibidores de Proteasas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Notch/efectos de los fármacos , Receptores Notch/genética , Telencéfalo/citología , Transfección
16.
Toxicol Sci ; 101(2): 310-20, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17977900

RESUMEN

Manganese is an essential nutrient for humans that has to be maintained at proper levels for normal brain functioning. However, manganese also acts as a toxicant to the brain, and several studies have linked exposure to excessive manganese to neurotoxicity in adults. A recent report has suggested that ingesting high doses of manganese via drinking water can impede intellectual functions in children. It is known that during development, the nervous system is particularly vulnerable to different types of injuries and toxicants. Neural stem cells (NSCs) play an essential role in both the developing nervous system and the adult brain where the capacity for self-renewal may be important. In the present study, we have used NSCs to investigate the molecular mechanisms involved in manganese developmental neurotoxicity. The results show that primary cultures of rat embryonic cortical NSCs as well as the murine-derived multipotent NSC line C17.2 undergo apoptotic cell death via a mitochondrial-mediated pathway in response to manganese. Exposed cells exhibit typical apoptotic features, such as chromatin condensation and cell shrinkage, mitochondrial cytochrome c release, activation of caspase-3, and caspase-specific cleavage of the endogenous substrate poly (ADP-ribose) polymerase. In addition, our data also show that reactive oxygen species formation plays a role in the onset of manganese toxicity in NSCs.


Asunto(s)
Apoptosis/efectos de los fármacos , Corteza Cerebral , Cloruros/toxicidad , Contaminantes Ambientales/toxicidad , Mitocondrias/efectos de los fármacos , Células Madre Multipotentes/efectos de los fármacos , Animales , Western Blotting , Calcio/metabolismo , Caspasas/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/patología , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Corteza Cerebral/patología , Citosol/efectos de los fármacos , Citosol/metabolismo , Citosol/patología , Compuestos de Manganeso , Ratones , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/patología , Orgánulos/efectos de los fármacos , Orgánulos/metabolismo , Orgánulos/patología , Especies Reactivas de Oxígeno/metabolismo
17.
Neurotox Res ; 34(4): 820-833, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29859004

RESUMEN

Pesticide exposure has been linked to the pathogenesis of neurodevelopmental and neurodegenerative disorders including autism spectrum disorders, attention deficit/hyperactivity, and Parkinson's disease (PD). Developmental exposure to pesticides, even at low concentrations not harmful for the adult brain, can lead to neuronal loss and functional deficits. It has been shown that prenatal or early postnatal exposure to the herbicide paraquat (PQ) and the fungicide maneb (MB), alone or in combination, causes permanent toxicity in the nigrostriatal dopamine system, supporting the idea that early exposure to these pesticides may contribute to the pathophysiology of PD. However, the mechanisms mediating PQ and MB developmental neurotoxicity are not yet understood. Therefore, we investigated the neurotoxic effect of low concentrations of PQ and MB in primary cultures of rat embryonic neural stem cells (NSCs), with particular focus on cell proliferation and oxidative stress. Exposure to PQ alone or in combination with MB (PQ + MB) led to a significant decrease in cell proliferation, while the cell death rate was not affected. Consistently, PQ + MB exposure altered the expression of major genes regulating the cell cycle, namely cyclin D1, cyclin D2, Rb1, and p19. Moreover, PQ and PQ + MB exposures increased the reactive oxygen species (ROS) production that could be neutralized upon N-acetylcysteine (NAC) treatment. Notably, in the presence of NAC, Rb1 expression was normalized and a normal cell proliferation pattern could be restored. These findings suggest that exposure to PQ + MB impairs NSCs proliferation by mechanisms involving alterations in the redox state.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Maneb/toxicidad , Células-Madre Neurales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Paraquat/toxicidad , Plaguicidas/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Relación Dosis-Respuesta a Droga , Herbicidas/toxicidad , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Estrés Oxidativo/fisiología , Ratas Sprague-Dawley
19.
EFSA J ; 16(1): e05083, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32625654

RESUMEN

The CONTAM Panel updated the assessment of the risks for human health related to the presence of 3-monochloropropane diol (3-MCPD) and its fatty acid esters in food published in 2016 in view of the scientific divergence identified in the establishment of the tolerable daily intake (TDI) in the Joint FAO/WHO Expert Committee on Food Additives and Contaminants (FAO/WHO) report published in 2017. In this update, dose-response analysis was performed following the recent EFSA Scientific Committee guidance on the use of benchmark dose (BMD) approach in risk assessment, and a review of available data on developmental and reproduction toxicity was included. The outcome of this review indicates that in rats short-term exposure to 3-MCPD above 1 mg/kg body weight (bw) per day can induce reduced sperm motility associated with reduced male fecundity. Decreased sperm count and histopathological changes in the testis and epididymis were observed following longer treatment periods at higher doses. Regarding increased incidence kidney tubular hyperplasia, BMD analysis using model averaging resulted in a BMDL 10 of 0.20 mg/kg bw per day in male rats, which was selected as the new Reference Point (RP) for renal effects. For the effects on male fertility, decreased sperm motility was selected as the most sensitive relevant endpoint and a BMDL 05 of 0.44 mg/kg bw per day was calculated. The RP for renal effects was considered to derive an updated group TDI of 2 µg/kg bw per day for 3-MCPD and its fatty acid esters and was considered protective also for effects on male fertility. The established TDI of 2 µg/kg bw per day is not exceeded in the adult population. A slight exceedance of the TDI was observed in the high consumers of the younger age groups and in particular for the scenarios on infants receiving formula only.

20.
EFSA J ; 16(12): e05194, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32625773

RESUMEN

The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) in food. Regarding PFOS and PFOA occurrence, the final data set available for dietary exposure assessment contained a total of 20,019 analytical results (PFOS n = 10,191 and PFOA n = 9,828). There were large differences between upper and lower bound exposure due to analytical methods with insufficient sensitivity. The CONTAM Panel considered the lower bound estimates to be closer to true exposure levels. Important contributors to the lower bound mean chronic exposure were 'Fish and other seafood', 'Meat and meat products' and 'Eggs and egg products', for PFOS, and 'Milk and dairy products', 'Drinking water' and 'Fish and other seafood' for PFOA. PFOS and PFOA are readily absorbed in the gastrointestinal tract, excreted in urine and faeces, and do not undergo metabolism. Estimated human half-lives for PFOS and PFOA are about 5 years and 2-4 years, respectively. The derivation of a health-based guidance value was based on human epidemiological studies. For PFOS, the increase in serum total cholesterol in adults, and the decrease in antibody response at vaccination in children were identified as the critical effects. For PFOA, the increase in serum total cholesterol was the critical effect. Also reduced birth weight (for both compounds) and increased prevalence of high serum levels of the liver enzyme alanine aminotransferase (ALT) (for PFOA) were considered. After benchmark modelling of serum levels of PFOS and PFOA, and estimating the corresponding daily intakes, the CONTAM Panel established a tolerable weekly intake (TWI) of 13 ng/kg body weight (bw) per week for PFOS and 6 ng/kg bw per week for PFOA. For both compounds, exposure of a considerable proportion of the population exceeds the proposed TWIs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA