Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Toxicol Appl Pharmacol ; 449: 116130, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35714712

RESUMO

Epidemiological studies have linked long-term/high-dose usage of paracetamol (N-acetyl-para-aminophenol, APAP) during pregnancy to adverse neuropsychiatric outcomes, primarily attention-deficit hyperactive disorder (ADHD), in the offspring. Though variable, ADHD has been associated with phenotypic alterations characterized by reductions in grey matter densities and aberrations in structural connectivity, effects which are thought to originate in neurodevelopment. We used embryonic chicken cerebellar granule neurons (CGNs) and neuronally differentiating human NTERA2 cells (NT2Ns) to investigate the in vitro effects of APAP on cell viability, migration, neuritogenesis, and the intracellular levels of various proteins involved in neurodevelopment as well as in the maintenance of the structure and function of neurites. Exposure to APAP ranging from 100 to 1600 µM yielded concentration- and time-dependent reductions in cell viability and levels of neurite arborization, as well as reductions in the levels of the cytoskeletal protein ß2-spectrin, with the highest APAP concentration resulting in between 50 and 75% reductions in the aforementioned metrics over the course of 72 h. Exposure to APAP also reduced migration in the NT2Ns but not CGNs. Moreover, we found concentration- and time-dependent increases in punctate aggregation of the cytoskeletal protein ß3-tubulin following exposure to APAP in both cell model systems, with the highest APAP concentration approximately doubling the number of aggregates over 72-120 h. Our findings demonstrate that APAP negatively perturbs neurite arborization degree, with concurrent reductions in the protein levels of ß2-spectrin and disruption of the integrity of ß3-tubulin, both proteins of which play important roles in neuronal structure and function.


Assuntos
Acetaminofen , Plasticidade Neuronal , Acetaminofen/efeitos adversos , Animais , Linhagem Celular , Embrião de Galinha , Proteínas do Citoesqueleto , Feminino , Humanos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Gravidez , Espectrina , Tubulina (Proteína)
2.
Neurochem Int ; 169: 105571, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37451345

RESUMO

Antidepressants are used to treat depression and some anxiety disorders, including use in pregnant patients. The pharmacological actions of these drugs generally determine the uptake and metabolism of a series of neurotransmitters, such as serotonin, norepinephrine, or dopamine, along with an increase in BDNF expression. However, many aspects of antidepressant action remain unknown, particularly whether antidepressants interfere with normal neurodevelopment when taken by pregnant women. In order to reveal cellular and molecular implications crucial to the functioning of pathways related to antidepressant effects, we performed an investigation on neuronally differentiating human SH-SY5Y cells. To our knowledge, this is the first time human SH-SY5Y cells in cultures of purely neuronal cells induced by controlled differentiation with retinoic acid are followed by short-term 48-h exposure to 0.1-10 µM escitalopram or venlafaxine. Treatment with antidepressants (1 µM) did not affect the electrophysiological properties of SH-SY5Y cells. However, the percentage of mature neurons exhibiting voltage-gated sodium currents was substantially higher in cultures pre-treated with either antidepressant. After exposure to escitalopram or venlafaxine, we observed a concentration-dependent increase in activity-dependent BDNF promoter IV activation. The assessment of neurite metrics showed significant down-regulation of neurite outgrowth upon exposure to venlafaxine. Identified changes may represent links to molecular processes of importance to depression and be involved in neurodevelopmental alterations observed in postpartum children exposed to antidepressants antenatally.


Assuntos
Escitalopram , Crescimento Neuronal , Cloridrato de Venlafaxina , Criança , Feminino , Humanos , Gravidez , Antidepressivos/farmacologia , Antidepressivos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Neuroblastoma/metabolismo , Crescimento Neuronal/efeitos dos fármacos , Neurônios/metabolismo , Cloridrato de Venlafaxina/farmacologia
3.
Neurotoxicol Teratol ; 90: 107057, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34942343

RESUMO

Exposing the immature nervous system to specific antiepileptic drugs (AEDs) during pregnancy is linked to neurodevelopmental disorders such as autism spectrum disorder (ASD). Newer AEDs like lamotrigine (LTG) are hailed as safer, but recent epidemiological data suggest that even LTG carries a risk, although much lower than that associated with valproic acid (VPA), an older AED, which is also known to cause morphological alterations in the developing brain. Increasing evidence highlights cerebellar abnormalities as important in ASD pathophysiology. Transcription factor PAX6 is a key activity-dependent mediator and regulates crucial processes during cerebellar development. The chicken cerebellum recapitulates important characteristics of human cerebellar development, and may thus be suitable for the assessment of interventions aiming to modify maturation and cerebellar development. In the present study, exposure of chicken on embryonic day 16 (E16) to LTG or VPA resulted in decreased cerebellar mass and level of proliferating nuclear cell antigen (PCNA) for clinically relevant concentrations of VPA. However, both AEDs reduced cerebellar protein levels of PAX6 and MMP-9 at E17. Furthermore, PAX6 immunohistochemical staining of coronal sections of chicken cerebellum showed a significant reduction in PAX6-positive cell density and changes in cerebellar cortex thickness, mostly caused by the change in IGL-layer thickness. In conclusion, prenatal exposure to LTG or VPA provoked differential maturational changes in the developing cerebellum that may reflect some of the underlying molecular mechanisms for the observed human ASD pathology after AEDs exposure during pregnancy.


Assuntos
Transtorno do Espectro Autista , Epilepsia , Animais , Anticonvulsivantes/toxicidade , Embrião de Galinha , Galinhas , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Feminino , Lamotrigina/uso terapêutico , Lamotrigina/toxicidade , Gravidez , Triazinas/uso terapêutico , Triazinas/toxicidade , Ácido Valproico/toxicidade
4.
Heliyon ; 8(8): e10291, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36051260

RESUMO

Chordoma associated with tuberous sclerosis complex (TSC) is an extremely rare tumor that was described only in 13 cases since 1975. Сhordoma itself is a malignant slow-growing bone tumor thought to arise from vestigial or ectopic notochordal tissue. Chordoma associated with TSC differs from chordoma in the general pediatric population in the median age, where the diagnosis of TSC-associated chordoma is 6.2 months, whereas for chordoma in the general pediatric population it is set to 12 years. The majority of TSC-associated chordomas are localized in skull-based and sacrum regions, and rare in the spine. Chordomas are genetically heterogeneous tumors characterized by chromosomal instability (CIN), and alterations involving PI3K-AKT signaling pathway genes and chromatin remodeling genes. Here we present the 14th case of chordoma associated with TSC in a 1-year-old pediatric patient. Alongside biallelic inactivation of the TSC1 gene, molecular genetic analysis revealed CIN and involvement of epigenetic regulation genes. In addition, we found the engagement of CBX7 and apolipoprotein B editing complex (APOBEC3) genes that were not yet seen in chordomas before. Amplification of CBX7 may epigenetically silence the CDKN2A gene, whereas amplification of APOBEC3 genes can explain the frequent occurrence of CIN in chordomas. We also found that KRAS gene is located in the region with gain status, which may suggest the ineffectiveness of potential EGFR monotherapy. Thus, molecular genetic analysis carried out in this study broadens the horizons of possible approaches for targeted therapies with potential applications for personalized medicine.

5.
J Pharmacol Toxicol Methods ; 112: 107105, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34284116

RESUMO

INTRODUCTION: Rodent models are routinely used to assess the safety and developmental toxicity of pharmaceuticals, along with analysis of their distribution. These models require sacrifice of parent females, have challenges in the estimation of the number of embryos and stage of development, and are expensive and time-consuming. In this study, we used fertilized chicken eggs as an alternative model to address drug distribution to the developing brain of two antiepileptic drugs, valproic acid (VPA) and lamotrigine (LTG) at two developmental stages. METHODS: VPA or LTG was injected into the allantois of the egg on embryonic day 13 (E13) or E16. Whole chicken brains were harvested at time-points of 5 min to 24 h and the concentrations of the drugs determined using GC/MS and LC-MS/MS, for VPA and LTG, respectively. RESULTS: VPA and LTG had distinct absorption and elimination phases and were found in the brain as early as 5-15 min after injection. Both drugs reached the brain in clinically relevant concentrations, with Cmax 10-30% of the calculated concentration assuming uniform distribution throughout the egg. LTG concentrations were higher when injected at E13 compared to E16. CONCLUSION: The chicken embryo model may be a suitable alternative animal model for preclinical drug distribution studies. It enables to easily approach antenatal development on an individual level, with a precise number of experimental animals, high reproducibility and low time and cost. Knowledge of the concentrations reaching the brain at different developmental stages with different drugs is important for the planning and interpretation of neurodevelopmental toxicity studies.


Assuntos
Epilepsia , Preparações Farmacêuticas , Animais , Anticonvulsivantes/uso terapêutico , Anticonvulsivantes/toxicidade , Encéfalo , Embrião de Galinha , Galinhas , Cromatografia Líquida , Modelos Animais de Doenças , Interações Medicamentosas , Epilepsia/tratamento farmacológico , Feminino , Gravidez , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem , Triazinas , Ácido Valproico/toxicidade
6.
Toxicol Lett ; 338: 85-96, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33309997

RESUMO

Disruption of neurite outgrowth is a marker for neurotoxicity. Persistent organic pollutants (POPs) are potential developmental neurotoxicants. We investigated their effect on neurite outgrowth in PC12 rat pheochromocytoma cells, in absence or presence of nerve growth factor (NGF), an inducer of neuronal differentiation. Cells were exposed for 72 h to a defined mixture of POPs with chemical composition and concentrations based on blood levels in the Scandinavian population. We also evaluated perfluorooctane sulfonic acid (PFOS) alone, the most abundant compound in the POP mixture. Only higher concentrations of POP mixture reduced tetrazolium salt (MTT) conversion. High-content analysis showed a decrease in cell number, but no changes for nuclear and mitochondrial cellular health parameters. Robust glutathione levels were observed in NGF-differentiated cells. Live imaging, using the IncuCyte ZOOM platform indicated ongoing cell proliferation over time, but slower in presence of NGF. The pollutants did not inhibit neuritogenesis, but rather increased NGF-induced neurite length. PFOS induced neurite outgrowth to about 50 % of the level seen with the POP mixture. Neither the POP mixture nor PFOS affected neurite length in the absence of NGF. Our observations indicate that realistic complex mixtures of environmental pollutants can affect neuronal connectivity via NGF-induced neurite outgrowth.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Crescimento Neuronal/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Neuritos/metabolismo , Neuritos/patologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Células PC12 , Ratos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA