Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Exp Biol Med (Maywood) ; 248(7): 633-640, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37208932

RESUMEN

Neurotoxicity assessments are generally performed using laboratory animals. However, as in vitro neurotoxicity models are continuously refined to reach adequate predicative concordance with in vivo responses, they are increasingly used for some endpoints of neurotoxicity. In this study, gestational day 80 fetal rhesus monkey brain tissue was obtained for neural stem cells (NSCs) isolation. Cells from the entire hippocampus were harvested, mechanically dissociated, and cultured for proliferation and differentiation. Immunocytochemical staining and biological assays demonstrated that the harvested hippocampal cells exhibited typical NSC phenotypes in vitro: (1) cells proliferated vigorously and expressed NSC markers nestin and sex-determining region Y-box 2 (SOX2) and (2) cells differentiated into neurons, astrocytes, and oligodendrocytes, as confirmed by positive staining with class III ß-tubulin, glial fibrillary acidic protein, and galactocerebroside, respectively. The NSC produced detectable responses following neurotoxicant exposures (e.g. trimethyltin and 3-nitropropionic acid). Our results indicated that non-human primate NSCs may be a practical tool to study the biology of neural cells and to evaluate the neurotoxicity of chemicals in vitro, thereby providing data that are translatable to humans and may also reduce the number of animals needed for developmental neurotoxicological studies.


Asunto(s)
Células-Madre Neurales , Animales , Neuronas/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Encéfalo
2.
J Magn Reson Imaging ; 56(5): 1499-1504, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35278003

RESUMEN

BACKGROUND: Currently, the gadolinium retention in the brain after the use of contrast agents is studied by T1 -weighted magnetic resonance imaging (MRI) (T1 w) and T1 mapping. The former does not provide easily quantifiable data and the latter requires prolonged scanning and is sensitive to motion. T2 mapping may provide an alternative approach. Animal studies of gadolinium retention are complicated by repeated intravenous (IV) dosing, whereas intraperitoneal (IP) injections might be sufficient. HYPOTHESIS: T2 mapping will detect the changes in the rat brain due to gadolinium retention, and IP administration is equivalent to IV for long-term studies. STUDY TYPE: Prospective longitudinal. ANIMAL MODEL: A total of 31 Sprague-Dawley rats administered gadodiamide IV (N = 8) or IP (N = 8), or saline IV (N = 6) or IP (N = 9) 4 days per week for 5 weeks. FIELD STRENGTH/SEQUENCES: A 7 T, T1 w, and T2 mapping. ASSESSMENT: T2 relaxation and image intensities in the deep cerebellar nuclei were measured pre-treatment and weekly for 5 weeks. Then brains were assessed for neuropathology (N = 4) or gadolinium content using inductively coupled plasma mass spectrometry (ICP-MS, N = 12). STATISTICAL TESTS: Repeated measures analysis of variance with post hoc Student-Newman-Keuls tests and Hedges' effect size. RESULTS: Gadolinium was detected by both approaches; however, T2 mapping was more sensitive (effect size 2.32 for T2 vs. 0.95 for T1 w), and earlier detection (week 3 for T2 vs. week 4 for T1 w). ICP-MS confirmed the presence of gadolinium (3.076 ± 0.909 nmol/g in the IV group and 3.948 ± 0.806 nmol/g in the IP group). There was no significant difference between IP and IV groups (ICP-MS, P = 0.109; MRI, P = 0.696). No histopathological abnormalities were detected in any studied animal. CONCLUSION: T2 relaxometry detects gadolinium retention in the rat brain after multiple doses of gadodiamide irrespective of the route of administration. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 1.


Asunto(s)
Medios de Contraste , Compuestos Organometálicos , Animales , Encéfalo/diagnóstico por imagen , Gadolinio/farmacología , Gadolinio DTPA , Imagen por Resonancia Magnética/métodos , Estudios Prospectivos , Ratas , Ratas Sprague-Dawley
3.
Int J Toxicol ; 40(4): 367-379, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33878910

RESUMEN

The organotin, trimethyltin (TMT), is a highly toxic compound. In this study, silver-stained rat brain sections were qualitatively and quantitatively evaluated for degeneration after systemic treatment with TMT. Degenerated neurons were counted using image analysis methods available in the HALO image analysis software. Specific brain areas including the cortex, inferior and superior colliculus, and thalamus were quantitatively analyzed. Our results indicate extensive and widespread damage to the rat brain after systemic administration of TMT. Qualitative results suggest severe TMT-induced toxicity 3 and 7 days after the administration of TMT. Trimethyltin toxicity was greatest in the hippocampus, olfactory area, cerebellum, pons, mammillary nucleus, inferior and superior colliculus, hypoglossal nucleus, thalamus, and cerebellar Purkinje cells. Quantification showed that the optic layer of the superior colliculus exhibited significantly more degeneration compared to layers above and below. The inferior colliculus showed greater degeneration in the dorsal area relative to the central area. Similarly, in cortical layers, there was greater neurodegeneration in deeper layers compared to superficial layers. Quantification of damage in various thalamic nuclei showed that the greatest degeneration occurred in midline and intralaminar nuclei. These results suggest selective neuronal network vulnerability to TMT-related toxicity in the rat brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Compuestos de Trimetilestaño/toxicidad , Animales , Encéfalo/patología , Masculino , Ratas , Ratas Sprague-Dawley
4.
J Appl Toxicol ; 41(7): 996-1006, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33140470

RESUMEN

Neurotoxicity studies are important in the preclinical stages of drug development process, because exposure to certain compounds that may enter the brain across a permeable blood brain barrier damages neurons and other supporting cells such as astrocytes. This could, in turn, lead to various neurological disorders such as Parkinson's or Huntington's disease as well as various dementias. Toxicity assessment is often done by pathologists after these exposures by qualitatively or semiquantitatively grading the severity of neurotoxicity in histopathology slides. Quantification of the extent of neurotoxicity supports qualitative histopathological analysis and provides a better understanding of the global extent of brain damage. Stereological techniques such as the utilization of an optical fractionator provide an unbiased quantification of the neuronal damage; however, the process is time-consuming. Advent of whole slide imaging (WSI) introduced digital image analysis which made quantification of neurotoxicity automated, faster and with reduced bias, making statistical comparisons possible. Although automated to a certain level, simple digital image analysis requires manual efforts of experts which is time-consuming and limits analysis of large datasets. Digital image analysis coupled with a deep learning artificial intelligence model provides a good alternative solution to time-consuming stereological and simple digital analysis. Deep learning models could be trained to identify damaged or dead neurons in an automated fashion. This review has focused on and discusses studies demonstrating the role of deep learning in segmentation of brain regions, toxicity detection and quantification of degenerated neurons as well as the estimation of area/volume of degeneration.


Asunto(s)
Inteligencia Artificial , Aprendizaje Profundo , Toxicología , Algoritmos , Encéfalo , Redes Neurales de la Computación
5.
Neurotoxicology ; 81: 172-179, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33045284

RESUMEN

We have described that prolonged sevoflurane exposure at a clinically-relevant concentration of 2.5 % caused neuronal cell death in the developing monkey brain. Postnatal day 5 or 6 rhesus monkeys (n = 3) were exposed to 2.5 % sevoflurane for 8 h. Monkeys kept at environmental conditions in the procedure room served as controls (n = 3). Brain tissues were harvested four hours after sevoflurane exposure for histological analysis, and RNA or protein extraction. MicroRNA (miRNA) profiling on the frontal cortex of monkey brains was performed using next-generation sequencing. 417 miRNAs were identified in the frontal cortex, where most neuronal cell death was observed. 7 miRNAs were differentially expressed in frontal cortex after sevoflurane exposure. Five of these were expressed at significantly lower levels than controls and the other two miRNAs were expressed significantly higher. These differentially expressed miRNAs (DEMs) were then loaded to the Ingenuity Pathway Analysis database for pathway analysis, in which targeting information was available for 5 DEMs. The 5 DEMs target 2,919 mRNAs which are involved in pathways that contribute to various cellular functions. Of note, 78 genes that are related to axon guidance signaling were targeted, suggesting that development of the neural circuit may be affected after sevoflurane exposure. Such changes may have long-term effects on brain development and function. These findings are supplementary to our previous observations and provide more evidence for better understanding the adverse effects of sevoflurane on the developing brain after an 8 -h exposure.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Encéfalo/efectos de los fármacos , Perfilación de la Expresión Génica , MicroARNs/genética , Neuronas/efectos de los fármacos , Sevoflurano/toxicidad , Transcriptoma/efectos de los fármacos , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Muerte Celular/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Macaca mulatta , Masculino , MicroARNs/metabolismo , Neuronas/metabolismo , Neuronas/patología , Mapas de Interacción de Proteínas , Factores de Tiempo
6.
Int J Toxicol ; 39(4): 294-306, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32468881

RESUMEN

This study consisted of a qualitative and quantitative assessment of neuropathological changes in kainic acid (KA)-treated adult male rats. Rats were administered a single 10 mg/kg intraperitoneal injection of KA or the same volume of saline and sacrificed 24 or 48 hours posttreatment. Brains were collected, sectioned coronally (∼ 81 slices), and stained with amino cupric silver to reveal degenerative changes. For qualitative assessment of neural degeneration, sectioned material was evaluated by a board-certified pathologist, and the level of degeneration was graded based upon a 4-point scale. For measurement of quantitative neural degeneration in response to KA treatment, the HALO digital image analysis software tool was used. Quantitative measurements of specific regions within the brain were obtained from silver-stained tissue sections with quantitation based on stain color and optical density. This quantitative evaluation method identified degeneration primarily in the cerebral cortex, septal nuclei, amygdala, olfactory bulb, hippocampus, thalamus, and hypothalamus. The KA-produced neuronal degeneration in the cortex was primarily in the piriform, insular, rhinal, and cingulate areas. In the hippocampus, the dentate gyrus was found to be the most affected area. Our findings indicate global neurotoxicity due to KA treatment. Certain brain structures exhibited more degeneration than others, reflecting differential sensitivity or vulnerability of neurons to KA.


Asunto(s)
Encéfalo/efectos de los fármacos , Ácido Kaínico/toxicidad , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad , Animales , Encéfalo/patología , Masculino , Neuronas/patología , Síndromes de Neurotoxicidad/patología , Ratas Sprague-Dawley
7.
Drug Metab Dispos ; 48(6): 447-458, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32193355

RESUMEN

Safety assessments of new drug candidates are an important part of the drug development and approval process. Often, possible sex-associated susceptibilities are not adequately addressed, and better assessment tools are needed. We hypothesized that hepatic transcript profiles of cytochrome P450 (P450) enzymes can be used to predict sex-associated differences in drug metabolism and possible adverse events. Comprehensive hepatic transcript profiles were generated for F344 rats of both sexes at nine ages, from 2 weeks (preweaning) to 104 weeks (elderly). Large differences in the transcript profiles of 29 drug metabolizing enzymes and transporters were found between adult males and females (8-52 weeks). Using the PharmaPendium data base, 41 drugs were found to be metabolized by one or two P450 enzymes encoded by sexually dimorphic mRNAs and thus were candidates for evaluation of possible sexually dimorphic metabolism and/or toxicities. Suspension cultures of primary hepatocytes from three male and three female adult rats (10-13 weeks old) were used to evaluate the metabolism of 11 drugs predicted to have sexually dimorphic metabolism. The pharmacokinetics of the drug or its metabolite was analyzed by liquid chromatography/tandem mass spectrometry using multiple reaction monitoring. Of those drugs with adequate metabolism, the predicted significant sex-different metabolism was found for six of seven drugs, with half-lives 37%-400% longer in female hepatocytes than in male hepatocytes. Thus, in this rat model, transcript profiles may allow identification of potential sex-related differences in drug metabolism. SIGNIFICANCE STATEMENT: The present study showed that sex-different expression of genes coding for drug metabolizing enzymes, specifically cytochrome P450s, could be used to predict sex-different drug metabolism and, thus, provide a new tool for protecting susceptible subpopulations from possible adverse drug events.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica , Tasa de Depuración Metabólica/genética , Animales , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Conjuntos de Datos como Asunto , Femenino , Perfilación de la Expresión Génica , Semivida , Hepatocitos , Hígado/enzimología , Masculino , Modelos Animales , Cultivo Primario de Células , Ratas , Ratas Endogámicas F344 , Factores Sexuales
8.
Neurotoxicology ; 77: 181-192, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32014511

RESUMEN

Bacterial cell wall endotoxins, i.e. lipopolysaccharides (LPS), are some of the original compounds shown to evoke the classic signs of systemic inflammation/innate immune response and neuroinflammation. The term neuroinflammation often is used to infer the elaboration of proinflammatory mediators by microglia elicited by neuronal targeted activity. However, it also is possible that the microglia are responding to vasculature through several signaling mechanisms. Microglial activation relative to the vasculature in the hippocampus and parietal cortex was determined after an acute exposure of a single subcutaneous injection of 2 mg/kg LPS. Antibodies to allograft inflammatory factor (Aif1, a.k.a. Iba1) were used to track and quantify morphological changes in microglia. Immunostaining of platelet/endothelial cell adhesion molecule 1 (Pecam1, a.k.a. Cd31) was used to visualize vasculature in the forebrain and glial acidic fibrillary protein (GFAP) to visualize astrocytes. Neuroinflammation and other aspects of neurotoxicity were evaluated histologically at 3 h, 6 h, 12 h, 24 h, 3 d and 14 d following LPS exposure. LPS did not cause neurodegeneration as determined by Fluoro Jade C labeling. Also, there were no signs of mouse IgG leakage from brain vasculature due to LPS. Some changes in microglia size occurred at 6 h, but by 12 h microglial activation had begun with the combined soma and proximal processes size increasing significantly (1.5-fold). At 24 h, almost all the microglia soma and proximal processes in the hippocampus, parietal cortex, and thalamus were closely associated with the vasculature and had increased almost 2.0-fold in size. In many areas where microglia were juxtaposed to vasculature, astrocytic endfeet appeared to be displaced. The microglial activation had subsided slightly by 3 d with microglial size 1.6-fold that of control. We hypothesize that acute LPS activation can result in vascular mediated microglial responses through several mechanisms: 1) binding to Cd14 and Tlr4 receptors on microglia processes residing on vasculature; 2) damaging vasculature and causing the release of cytokines; and 3) possibly astrocytic endfeet damage resulting in cytokine release. These acute responses may serve as an adaptive mechanism to exposure to circulating LPS where the microglia surround the vasculature. This could further prevent the pathogen(s) circulating in blood from entering the brain. However, diverting microglial interactions away from synaptic remodeling and other types of microglial interactions with neurons may have adverse effects on neuronal function.


Asunto(s)
Encefalitis/inmunología , Hipocampo/irrigación sanguínea , Hipocampo/inmunología , Lipopolisacáridos/toxicidad , Microglía/inmunología , Corteza Prefrontal/irrigación sanguínea , Corteza Prefrontal/inmunología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Encefalitis/inducido químicamente , Femenino , Hipocampo/efectos de los fármacos , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos
9.
Mol Neurobiol ; 57(1): 217-225, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31522383

RESUMEN

Numerous studies suggest a long duration of anesthesia during the late gestation period and infancy is associated with an increased risk of neuronal damage and neurocognitive impairment. The noble gas xenon is an anesthetic that is reported to have neuroprotective effects in some circumstances at certain concentrations. Currently, the effects of xenon on the brain and its potential neuroprotective properties, and/or the effects of xenon used in combination with other anesthetics, are not clearly understood and some reported data appear contradictory. In the present study, human neural stem cells were employed as a human-relevant model to evaluate the effects of xenon when it was co-administered with propofol, a frequently used anesthetic in pediatric anesthesia, and to understand the mechanism(s). The expression of polysialic acid (PSA) neural cell adhesion molecule (NCAM) on human neural stem cell-differentiated neurons was investigated as a key target molecule. PSA is a specific marker of developing neurons. It is essential for neuronal viability and plasticity. Human neural stem cells were maintained in neural differentiation medium and directed to differentiate into neuronal and glial lineages, and were exposed to propofol (50 µM) for 16 h in the presence or absence of xenon (33%). The neural stem cell-derived neurons were characterized by labelling cells with PSA-NCAM, after 5 days of differentiation. Propofol- and/or xenon-induced neurotoxicities were determined by measuring PSA immunoreactivity. A time course study showed that neuronal cell surface PSA was clearly cleaved off from NCAM by endoneuraminidase N (Endo-N), and eliminated PSA immunostaining was not re-expressed 4, 8, or 16 h after Endo-N washout. However, in the presence of 33% xenon, intense PSA staining on neuronal cell surface and processes was evident 16 h after Endo-N washout. In addition, prolonged (16 h) propofol exposure significantly decreased the positive rate of PSA-labeled neurons. When combined with xenon, propofol's adverse effects on neurons were attenuated. This work, conducted on the human neural stem cell-derived models, has provided evidence of the beneficiary effects of xenon on neurons and helps develop xenon-based anesthesia regimens in the pediatric population.


Asunto(s)
Anestésicos/farmacología , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Células-Madre Neurales/citología , Neuronas/metabolismo , Ácidos Siálicos/metabolismo , Xenón/farmacología , Células Cultivadas , Humanos , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Factores de Tiempo
10.
Mol Neurobiol ; 57(1): 200-207, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31578707

RESUMEN

Early life exposure to general anesthetics can have neurotoxic consequences. Evidence indicates that xenon, a rare noble gas with anesthetic properties, may lessen neuronal damage under certain conditions. However, its potential neuroprotective properties, when used alone or in combination with other anesthetics, remain largely unknown. While it is difficult to verify the adverse effects of long duration anesthetic exposure in infants and children, the utilization of relevant non-clinical models (i.e., human-derived neural stem cells) may serve as a "bridging" model for evaluating the vulnerability of the nervous system. Neural stem cells, purchased from PhoenixSongs Biologicals, Inc., were guided to differentiate into neurons, astrocytes, and oligodendrocytes, which were then exposed to propofol (50 µM) for 16 h in the presence or absence of xenon (33%). Differentiation into cells of the neural lineage was confirmed by labelling with cell-specific markers, ß-tubulin for neurons, glial fibrillary acidic protein (GFAP) for astrocytes, and galactocerebroside (GALC) for oligodendrocytes after 5 days of differentiation. The presence and severity of neural damage induced by anesthetic exposures were assessed by several methods, including the TUNEL assay, and immuno-histochemical measurements. Our data demonstrate that prolonged exposure to propofol results in a significant increase in the number of TUNEL-positive cells, indicating increased neural apoptosis. No significant changes were detected in the number of GFAP-positive astrocytes or GALC-positive oligodendrocytes. However, the number of ß-tubulin-positive neurons was substantially reduced in the propofol-exposed cultures. Co-administration of xenon effectively blocked the propofol-induced neuronal damage/loss. No significant effects were observed when xenon was administered alone. The data indicate that prolonged exposure to propofol during development produces elevated levels of neuronal apoptosis in a human neural stem cell-derived model. However, sub-clinical, non-anesthetic concentrations of xenon, when used in combination with propofol, can prevent or ameliorate the toxic effects associated with prolonged anesthetic exposure. This is important as a more complete understanding of the neurotoxic mechanisms associated with a variety of clinically relevant anesthetic combinations becomes available. Protective approaches are critical for developing sound guidance on best practices for the use of these agents in the pediatric setting.


Asunto(s)
Astrocitos/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Xenón/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Propofol/farmacología
11.
PLoS One ; 14(2): e0210273, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30779732

RESUMEN

This work extends the understanding of how toxic exposures to amphetamine (AMPH) adversely affect the immune system and lead to tissue damage. Importantly, it determines which effects of AMPH are and are not due to pronounced hyperthermia. Whole blood messenger RNA (mRNA) and whole blood and serum microRNA (miRNA) transcripts were identified in adult male Sprague-Dawley rats after exposure to toxic AMPH under normothermic conditions, AMPH when it produces pronounced hyperthermia, or environmentally-induced hyperthermia (EIH). mRNA transcripts with large increases in fold-change in treated relative to control rats and very low expression in the control group were a rich source of organ-specific transcripts in blood. When severe hyperthermia was produced by either EIH or AMPH, significant increases in circulating organ-specific transcripts for liver (Alb, Fbg, F2), pancreas (Spink1), bronchi/lungs (F3, Cyp4b1), bone marrow (Np4, RatNP-3b), and kidney (Cesl1, Slc22a8) were observed. Liver damage was suggested also by increased miR-122 levels in the serum. Increases in muscle/heart-enriched transcripts were produced by AMPH even in the absence of hyperthermia. Expression increases in immune-related transcripts, particularly Cd14 and Vcan, indicate that AMPH can activate the innate immune system in the absence of hyperthermia. Most transcripts specific for T-cells decreased 50-70% after AMPH exposure or EIH, with the noted exception of Ccr5 and Chst12. This is probably due to T-cells leaving the circulation and down-regulation of these genes. Transcript changes specific for B-cells or B-lymphoblasts in the AMPH and EIH groups ranged widely from decreasing ≈ 40% (Cd19, Cd180) to increasing 30 to 100% (Tk1, Ahsa1) to increasing ≥500% (Stip1, Ackr3). The marked increases in Ccr2, Ccr5, Pld1, and Ackr3 produced by either AMPH or EIH observed in vivo provide further insight into the initial immune system alterations that result from methamphetamine and AMPH abuse and could modify risk for HIV and other viral infections.


Asunto(s)
Trastornos Relacionados con Anfetaminas/sangre , Anfetamina/administración & dosificación , Fiebre/sangre , Golpe de Calor/sangre , MicroARNs/sangre , ARN Mensajero/sangre , Anfetamina/farmacología , Animales , Biomarcadores/sangre , Fiebre/inducido químicamente , Masculino , Ratas , Ratas Sprague-Dawley
12.
Neurotoxicology ; 70: 80-90, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30445043

RESUMEN

Despite the widespread use of general anesthesia, a growing body of research suggests that anesthesia exposure early in life may be associated with acute neurotoxicity and lasting behavioral changes. To better evaluate the risk posed by early life anesthesia on cognitive development, infant rhesus monkeys were exposed to an anesthesia regimen previously shown to be neurotoxic and their cognitive development was subsequently measured using a translational operant test battery. On postnatal day 5 or 6, animals were exposed to 8 h of isoflurane (n = 6, 1% isoflurane in a vehicle gas of 70% nitrous oxide and 30% oxygen) or a control condition (n = 8). Starting at 7 months of age, the monkeys were continuously trained and assessed on the NCTR Operant Test Battery (OTB). The OTB consists of cognitive tests which also exist in near identical forms for use in rats and humans, and includes tests of learning, memory, color discrimination, and motivation. Monkeys previously exposed to anesthesia showed a clear decrease in responding in a measure of motivation, as well as a lower response rate in a learning task. These data further support the hypothesis that prolonged anesthesia early in life may increase the risk of developing cognitive impairments later in life.


Asunto(s)
Anestesia General/efectos adversos , Cognición/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Isoflurano/toxicidad , Óxido Nitroso/toxicidad , Anestesia General/tendencias , Animales , Cognición/fisiología , Condicionamiento Operante/fisiología , Femenino , Isoflurano/administración & dosificación , Macaca mulatta , Masculino , Óxido Nitroso/administración & dosificación , Primates
13.
Neurotoxicology ; 69: 130-140, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30282018

RESUMEN

The initial goals of these experiments were to determine: 1) if blood-brain barrier (BBB) breakdown was a cause or an effect of METH-induced seizures; 2) all the brain regions where BBB is disrupted as seizures progress; and 3) the correlations between body temperature and vascular leakage and neurodegeneration. A fourth objective was added after initial experimentation to determine if sub-strain differences existed in adult male C57 B6 J (Jackson laboratories, JAX) versus C57 B6N (Charles River, CR) mice involving their susceptibility to BBB breakdown and seizure severity. With the 1st "maximal" intensity myoclonic-tonic seizure (MCT) varying degrees of IgG infiltration across the BBB (≤1 mm2) were prominent in olfactory system (OS) associated regions and in thalamus, hypothalamus and neocortex. IgG infiltration areas in the OS-associated regions of the bed nucleus of the stria terminalis, septum and more medial amygdala nuclei, and the hypothalamus were increased significantly by the time continuous behavioral seizures (CBS) developed. Mice receiving METH that had body temperatures of ≥40 °C had IgG infiltration along with MCT or CBS but peak body temperatures above 40 °C did not significantly increase IgG infiltration. Neurodegeneration seen at ≥6 h was restricted to the OS in both JAX and CR mice and was most prominent in the posteromedial cortical amygdaloid nucleus. Neurodegeneration in the anterior septum (tenia tecta) was seen only in the JAX mice. We hypothesize that METH-induced hypertension and hyperthermia lead to BBB breakdown and other vascular dysfunctions in the OS brain regions resulting in OS hyperexcitation. Excitation of the OS neural network then leads to the development of seizures. These seizures in turn exacerbate the energy depletions and the reactive oxygen stress produced by hyperthermia further damaging the BBB and vascular function. These events form a recurrent cycle that results in ever increasing seizure activity and neurotoxicity.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar/fisiología , Estimulantes del Sistema Nervioso Central/toxicidad , Progresión de la Enfermedad , Metanfetamina/toxicidad , Convulsiones/sangre , Convulsiones/inducido químicamente , Animales , Permeabilidad Capilar/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Convulsiones/diagnóstico , Factores de Tiempo
14.
Anal Chim Acta ; 1037: 87-96, 2018 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-30292318

RESUMEN

Although numerous studies have raised public concerns regarding the safety of anesthetics including sevoflurane in children, the biochemical mechanisms leading to anesthetics-induced neurotoxicity remain elusive. Moreover, potential biomarker(s) for early detection of general anesthetics-induced brain injury are urgent for public health. We employed an enabling technology of shotgun lipidomics and analyzed nearly 20 classes and subclasses of lipids present in the blood serum of postnatal day (PND) 5 or 6 rhesus monkeys temporally collected after exposure to sevoflurane at a clinically relevant concentration or room-air as control. Lipidomics analysis revealed numerous significant anesthetic-induced changes of serum lipids and their metabolites as well as short chain acylcarnitines in the brain and cerebrospinal fluid after anesthetic exposure. These include decreased carnitine and acylcarnitines, unchanged triacylglycerol mass but accumulation of 16:0 and 18:1 fatty acyl chains in the triacylglycerol pool, losses of polyunsaturated fatty acids in both non-esterified fatty acid and phospholipid pools, and increased 4-hydroxynonenal content as early as 2 h after sevoflurane exposure. Importantly, the amounts of short chain acylcarnitines in the brain and cerebrospinal fluid were also significantly reduced after anesthetic exposure. We propose that this serum lipidomic profile can serve as indicative of neuronal damage. Our results reveal that sevoflurane exposure induces an energy deficient state in the brain evidenced by reduced free and acyl carnitine contents, as well as the presence of a pro-inflammatory state in the exposed animals, providing deep insights into the underlying mechanisms responsible for anesthetic-induced neurotoxicity.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Neuronas/efectos de los fármacos , Sevoflurano/farmacología , Animales , Macaca mulatta
15.
Toxicol Appl Pharmacol ; 354: 191-195, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29550513

RESUMEN

Adverse effects related to central nervous system (CNS) function in pediatric populations may, at times, be difficult, if not impossible to evaluate. Prolonged anesthetic exposure affects brain excitability and anesthesia during the most sensitive developmental stages and has been associated with mitochondrial dysfunction, aberrant lipid metabolism and synaptogenesis, subsequent neuronal damage, as well as long-term behavioral deficits. There has been limited research evaluating whether and how anesthetic agents affect cellular lipids, the most abundant components of the brain other than water. Therefore, this review discusses: (1) whether the observed anesthetic-induced changes in lipid profiles seen in preclinical studies represents early signs of neurotoxicity; (2) the potential mechanisms underlying anesthetic-induced brain injury; and (3) whether lipid biomarker(s) identified in preclinical studies can serve as markers for the early clinical detection of anesthetic-induced neurotoxicity.


Asunto(s)
Anestesia/efectos adversos , Anestésicos/efectos adversos , Encéfalo/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/sangre , Metabolómica/métodos , Síndromes de Neurotoxicidad/etiología , Adolescente , Factores de Edad , Animales , Biomarcadores/sangre , Encéfalo/metabolismo , Encéfalo/fisiopatología , Niño , Desarrollo Infantil/efectos de los fármacos , Preescolar , Diagnóstico Precoz , Humanos , Lactante , Recién Nacido , Espectrometría de Masas , Síndromes de Neurotoxicidad/sangre , Síndromes de Neurotoxicidad/diagnóstico , Síndromes de Neurotoxicidad/fisiopatología , Valor Predictivo de las Pruebas , Factores de Riesgo
16.
Neurotoxicology ; 65: 98-110, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29427613

RESUMEN

Thiamine/vitamin B1 deficiency can lead to behavioral changes and neurotoxicity in humans. This may due in part to vascular damage, neuroinflammation and neuronal degeneration in the diencephalon, which is seen in animal models of pyrithiamine-enhanced thiamine deficiency. However, the time course of the progression of these changes in the animal models has been poorly characterized. Therefore, in this study, the progression of: 1) activated microglial association with vasculature; 2) neurodegeneration; and 3) any vascular leakage in the forebrain during the progress of thiamine deficiency were determined. A thiamine deficient diet along with 0.25 mg/kg/d of pyrithiamine was used as the mouse model. Vasculature was identified with Cd31 and microglia with Cd11b and Iba1 immunoreactivity. Neurodegeneration was determined by FJc labeling. The first sign of activated microglia within the thalamic nuclei were detected after 8 d of thiamine deficiency, and by 9 d activated microglia associated primarily with vasculature were clearly present but only in thalamus. At the 8 d time point neurodegeneration was not present in thalamus. However at 9 d, the first signs of neurodegeneration (FJc + neurons) were seen in most animals. Over 80% of the microglia were activated at 10 d but almost exclusively in the thalamus and the number of degenerating neurons was less than 10% of the activated microglia. At 10 d, there were sporadic minor changes in IgG presence in thalamus indicating minor vascular leakage. Dizocilpine (0.2-0.4 mg/kg) or phenobarbital (10-20 mg/kg) was administered to groups of mice from day 8 through day 10 to block neurodegeneration but neither did. In summary, activated microglia start to surround vasculature 1-2 d prior to the start of neurodegeneration. This response may be a means of controlling or repairing vascular damage and leakage. Glutamate excitotoxicity and vascular leakage likely only play a minor role in the early neurodegeneration resulting from thiamine deficiency. However, failure of dysfunctional vasculature endothelium to supply sufficient nutrients to neurons could be contributing to the neurodegeneration.


Asunto(s)
Vasos Sanguíneos/patología , Microglía/metabolismo , Degeneración Nerviosa/patología , Tálamo/metabolismo , Tálamo/patología , Deficiencia de Tiamina/metabolismo , Deficiencia de Tiamina/patología , Animales , Antígeno CD11b/metabolismo , Proteínas de Unión al Calcio/metabolismo , Dieta , Maleato de Dizocilpina/farmacología , Femenino , Ratones , Proteínas de Microfilamentos/metabolismo , Degeneración Nerviosa/prevención & control , Fenobarbital/farmacología , Piritiamina , Deficiencia de Tiamina/inducido químicamente , Factores de Tiempo
17.
Exp Biol Med (Maywood) ; 243(3): 228-236, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29105512

RESUMEN

Neurotoxicity has been linked with exposure to a number of common drugs and chemicals, yet efficient, accurate, and minimally invasive methods to detect it are lacking. Fluid-based biomarkers such as those found in serum, plasma, urine, and cerebrospinal fluid have great potential due to the relative ease of sampling but at present, data on their expression and translation are lacking or inconsistent. In this pilot study using a trimethyl tin rat model of central nervous system toxicity, we have applied state-of-the-art assessment techniques to identify potential individual biomarkers and patterns of biomarkers in serum, plasma, urine or cerebral spinal fluid that may be indicative of nerve cell damage and degeneration. Overall changes in metabolites and microRNAs were observed in biological fluids that were associated with neurotoxic damage induced by trimethyl tin. Behavioral changes and magnetic resonance imaging T2 relaxation and ventricle volume changes served to identify animals that responded to the adverse effects of trimethyl tin. Impact statement These data will help design follow-on studies with other known neurotoxicants to be used to assess the broad applicability of the present findings. Together this approach represents an effort to begin to develop and qualify a set of translational biochemical markers of neurotoxicity that will be readily accessible in humans. Such biomarkers could prove invaluable for drug development research ranging from preclinical studies to clinical trials and may prove to assist with monitoring of the severity and life cycle of brain lesions.


Asunto(s)
Biomarcadores , Líquidos Corporales/química , Sistema Nervioso Central/patología , MicroARNs/análisis , Neuronas/patología , Síndromes de Neurotoxicidad/diagnóstico , Compuestos de Trimetilestaño/toxicidad , Aminoácidos/análisis , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/orina , Humanos , Imagen por Resonancia Magnética , Masculino , Metaboloma/fisiología , MicroARNs/genética , Proyectos Piloto , Ratas , Ratas Sprague-Dawley
18.
J Neurochem ; 143(2): 198-213, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28792619

RESUMEN

Our previous studies have raised the possibility that altered blood glucose levels may influence and/or be predictive of methamphetamine (METH) neurotoxicity. This study evaluated the effects of exogenous glucose and corticosterone (CORT) pretreatment alone or in combination with METH on blood glucose levels and the neural and vascular toxicity produced. METH exposure consisted of four sequential injections of 5, 7.5, 10, and 10 mg/kg (2 h between injections) D-METH. The three groups given METH in combination with saline, glucose (METH+Glucose), or CORT (METH+CORT) had significantly higher glucose levels compared to the corresponding treatment groups without METH except at 3 h after the last injection. At this last time point, the METH and METH+Glucose groups had lower levels than the non-METH groups, while the METH+CORT group did not. CORT alone or glucose alone did not significantly increase blood glucose. Mortality rates for the METH+CORT (40%) and METH+Glucose (44%) groups were substantially higher than the METH (< 10%) group. Additionally, METH+CORT significantly increased neurodegeneration above the other three METH treatment groups (≈ 2.5-fold in the parietal cortex). Thus, maintaining elevated levels of glucose during METH exposure increases lethality and may exacerbate neurodegeneration. Neuroinflammation, specifically microglial activation, was associated with degenerating neurons in the parietal cortex and thalamus after METH exposure. The activated microglia in the parietal cortex were surrounding vasculature in most cases and the extent of microglial activation was exacerbated by CORT pretreatment. Our findings show that acute CORT exposure and elevated blood glucose levels can exacerbate METH-induced vascular damage, neuroinflammation, neurodegeneration and lethality. Cover Image for this issue: doi. 10.1111/jnc.13819.


Asunto(s)
Glucemia/efectos de los fármacos , Corticosterona/toxicidad , Glucosa/toxicidad , Metanfetamina/toxicidad , Lóbulo Parietal/efectos de los fármacos , Tálamo/efectos de los fármacos , Animales , Glucemia/metabolismo , Corticosterona/administración & dosificación , Combinación de Medicamentos , Glucosa/administración & dosificación , Masculino , Metanfetamina/administración & dosificación , Microglía/efectos de los fármacos , Microglía/metabolismo , Lóbulo Parietal/irrigación sanguínea , Lóbulo Parietal/metabolismo , Ratas , Ratas Sprague-Dawley , Tálamo/irrigación sanguínea , Tálamo/metabolismo
19.
Neurotoxicology ; 57: 298-309, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27984051

RESUMEN

Prolonged vitamin B1 (thiamine) deficiency can lead to neurological disorders such as Wernicke's encephalopathy and Wernicke-Korsakoff Syndrome (WKS) in humans. These thiamine deficiency disorders have been attributed to vascular leakage, blood-brain barrier breakdown and neuronal loss in the diencephalon and brain stem. However, endothelial dysfunction following thiamine deficiency and its relationship to the phenomenon of neurodegeneration has not been clearly elucidated. The present study sought to begin to address this issue by evaluating vascular morphology and integrity in a pyrithiamine (PT)-induced rat model of thiamine deficiency. Adjacent brain sections were used to either assess vascular integrity through immunohistochemical localization of rat endothelial cell antigen (RECA-1) and endothelial brain barrier antigen (EBA-1) or neurodegeneration using the de Olmos cupric silver method. GFAP and CD11b immunolabeling was used to evaluate astrocytic and microglial/macrophagic changes. Extensive neurodegeneration occurred concomitant with both vascular damage (thinning and breakage) and microglial activation in the inferior olive, medial thalamic area, and medial geniculate nuclei of pyrithiamine treated rats. Likewise, glucose transporter-1 (Glut-1), which is mostly expressed in endothelial cells, was also severely decreased in this pyrithiamine induced thiamine deficient rat model. MRI scans of these animals prior to sacrifice show that the pyrithiamine induced thiamine deficient animals have abnormal T2 relaxation values, which are commensurate with, and possibly predictive of, the neurodegeneration and/or endothelial dysfunction subsequently observed histologically in these same animals.


Asunto(s)
Antimetabolitos/toxicidad , Encéfalo/patología , Células Endoteliales/efectos de los fármacos , Piritiamina/toxicidad , Deficiencia de Tiamina/inducido químicamente , Deficiencia de Tiamina/patología , Animales , Antígenos de Superficie/metabolismo , Astrocitos/patología , Astrocitos/ultraestructura , Encéfalo/diagnóstico por imagen , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/ultraestructura , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Imagen por Resonancia Magnética , Masculino , Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/etiología , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata , Deficiencia de Tiamina/complicaciones , Deficiencia de Tiamina/diagnóstico por imagen
20.
Neurotoxicology ; 56: 225-232, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27555423

RESUMEN

Longitudinal MRI employing diffusion tensor imaging and T2 mapping approaches has been applied to investigate the mechanisms of white matter damage caused by acute hexachlorophene neurotoxicity in rats in vivo. Male Sprague-Dawley rats were administered hexachlorophene orally once a day for five consecutive days at a dose of 30mg/kg and were monitored in 7T MRI scanner at days 0 (baseline), 3, 6, 13, and 20 following the first hexachlorophene dose. Quantitative T2 maps as well as a number of diffusion tensor parameters (fractional anisotropy, radial and axial diffusivity, apparent diffusion coefficient, and trace) were calculated from corresponding MR images. T2, as well as all diffusion tensor derived parameters (except fractional anisotropy) showed significant changes during the course of neurotoxicity development. These changes peaked at 6days after the first dose of hexachlorophene (one day after the last dose) and recovered to practically baseline levels at the end of observation (20days from the first dose). While such changes in diffusivity and T2 relaxation clearly demonstrate myelin perturbations consistent with edema, the lack of changes of fractional anisotropy suggests that the structure of the myelin sheath was not disrupted significantly by hexachlorophene in this study. This is also confirmed by the rapid recovery of all observed MRI parameters after cessation of hexachlorophene exposure.


Asunto(s)
Antiinfecciosos Locales/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Imagen de Difusión Tensora , Hexaclorofeno/farmacología , Animales , Anisotropía , Procesamiento de Imagen Asistido por Computador , Estudios Longitudinales , Masculino , Fibras Nerviosas Mielínicas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...