Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Cell Biol Toxicol ; 40(1): 51, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38958792

RESUMO

The implementation of Zinc oxide nanoparticles (ZnO NPs) raises concerns regarding their potential toxic effects on human health. Although more and more researches have confirmed the toxic effects of ZnO NPs, limited attention has been given to their impact on the early embryonic nervous system. This study aimed to explore the impact of exposure to ZnO NPs on early neurogenesis and explore its underlying mechanisms. We conducted experiments here to confirm the hypothesis that exposure to ZnO NPs causes neural tube defects in early embryonic development. We first used mouse and chicken embryos to confirm that ZnO NPs and the Zn2+ they release are able to penetrate the placental barrier, influence fetal growth and result in incomplete neural tube closure. Using SH-SY5Y cells, we determined that ZnO NPs-induced incomplete neural tube closure was caused by activation of various cell death modes, including ferroptosis, apoptosis and autophagy. Moreover, dissolved Zn2+ played a role in triggering widespread cell death. ZnO NPs were accumulated within mitochondria after entering cells, damaging mitochondrial function and resulting in the over production of reactive oxygen species, ultimately inducing cellular oxidative stress. The N-acetylcysteine (NAC) exhibits significant efficacy in mitigating cellular oxidative stress, thereby alleviating the cytotoxicity and neurotoxicity brought about by ZnO NPs. These findings indicated that the exposure of ZnO NPs in early embryonic development can induce cell death through oxidative stress, resulting in a reduced number of cells involved in early neural tube closure and ultimately resulting in incomplete neural tube closure during embryo development. The findings of this study could raise public awareness regarding the potential risks associated with the exposure and use of ZnO NPs in early pregnancy.


Assuntos
Desenvolvimento Embrionário , Defeitos do Tubo Neural , Tubo Neural , Estresse Oxidativo , Espécies Reativas de Oxigênio , Óxido de Zinco , Óxido de Zinco/toxicidade , Animais , Estresse Oxidativo/efeitos dos fármacos , Embrião de Galinha , Desenvolvimento Embrionário/efeitos dos fármacos , Camundongos , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Tubo Neural/metabolismo , Humanos , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/patologia , Espécies Reativas de Oxigênio/metabolismo , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Feminino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nanopartículas Metálicas/toxicidade , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Nanopartículas/toxicidade
2.
Toxicol Appl Pharmacol ; 489: 117009, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38906509

RESUMO

INTRODUCTION: Aripiprazole (ARI) is a recently developed antipsychotic medication that belongs to the second generation of antipsychotics. The literature has contradictory information regarding ARI, which has been classified as pregnant use category C by the FDA. METHODS: 125 pathogen-free fertilized eggs were incubated for 28 h and divided into five groups of 25 eggs each (including the control group), and 18 eggs with intact integrity were selected from each group. After the experimental groups were divided, ARI was administered subblastodermally with a Hamilton micro-injector at 4 different doses (1 mg/kg, 5 mg/kg, 10 mg/kg, 20 mg/kg). At the 48th hour of incubation, all eggs were hatched and embryos were removed from the embryonic membranes. And then morphologic (position of the neural tube (open or closed), crown-rump length, number of somites, embryological development status), histopathologic (apoptosis (caspase 3), cell proliferation (PCNA), in situ recognition of DNA breaks (tunnel)), genetic (BRE gene expression) analyzes were performed. RESULTS: According to the results of the morphological analysis, when the frequency of neural tube patency was evaluated among the experimental groups, a statistically significant difference was determined between the control group and all groups (p < 0.001). In addition, the mean crown-rump length and somite number of the embryos decreased in a dose-dependent manner compared to the control group. It was determined that mRNA levels of the BRE gene decreased in embryos exposed to ARI compared to the control group (p < 0.001). CONCLUSION: Morphologically, histopathologically, and genetically, aripiprazole exposure delayed neurogenesis and development in early chick embryos. These findings suggest its use in pregnant women may be teratogenic. We note that these results are preliminary for pregnant women, but they should be expanded and studied with additional and other samples.


Assuntos
Aripiprazol , Tubo Neural , Animais , Aripiprazol/toxicidade , Tubo Neural/efeitos dos fármacos , Embrião de Galinha , Antipsicóticos/toxicidade , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Caspase 3/metabolismo , Caspase 3/genética
3.
Toxicol Appl Pharmacol ; 489: 117011, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38906510

RESUMO

The critical developmental stages of the embryo are strongly influenced by the dietary composition of the mother. Acrylamide is a food contaminant that can form in carbohydrate-rich foods that are heat-treated. The aim of this study was to investigate the toxicity of a relatively low dose of acrylamide on the development of the neural tube in the early stage chick embryos. Specific pathogen-free fertilized eggs (n = 100) were treated with acrylamide (0.1, 0.5, 2.5, 12.5 mg/kg) between 28-30th hours of incubation and dissected at 48th hours. In addition to morphological and histopathological examinations, proliferating cell nuclear antigen (PCNA) and caspase 3 were analyzed immunohistochemically. The brain and reproductive expression gene (BRE) was analyzed by RT-PCR. Acrylamide exposure had a negative effect on neural tube status even at a very low dose (0.1 mg/kg) (p < 0.05). Doses of 0.5 mg/kg and above caused a delay in neural tube development (p < 0.05). Crown-rump length and somite count decreased dose-dependently, while this decrease was not significant in the very low dose group (p > 0.05), which was most pronounced at doses of 2.5 and 12.5 mg/kg (p < 0.001). Acrylamide exposure dose-dependently decreased PCNA and increased caspase 3, with this change being significant at doses of 0.5 mg/kg and above (p < 0.001). BRE was downregulated at all acrylamide exposures except in the very low dose group (0.1 mg/kg). In conclusion, we find that acrylamide exposure (at 0.5 mg/kg and above) in post-gastrulation delays neural tube closure in chicken embryos by suppressing proliferation and apoptosis induction and downregulating BRE gene expression.


Assuntos
Acrilamida , Relação Dose-Resposta a Droga , Desenvolvimento Embrionário , Antígeno Nuclear de Célula em Proliferação , Animais , Embrião de Galinha , Acrilamida/toxicidade , Antígeno Nuclear de Célula em Proliferação/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Caspase 3/metabolismo , Caspase 3/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos
4.
Int J Mol Sci ; 25(11)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38891776

RESUMO

Neural tube defects (NTDs), which are caused by impaired embryonic neural tube closure, are one of the most serious and common birth defects. Peptidyl-prolyl cis/trans isomerase 1 (Pin1) is a prolyl isomerase that uniquely regulates cell signaling by manipulating protein conformation following phosphorylation, although its involvement in neuronal development remains unknown. In this study, we explored the involvement of Pin1 in NTDs and its potential mechanisms both in vitro and in vivo. The levels of Pin1 expression were reduced in NTD models induced by all-trans retinoic acid (Atra). Pin1 plays a significant role in regulating the apoptosis, proliferation, differentiation, and migration of neurons. Moreover, Pin1 knockdown significantly was found to exacerbate oxidative stress (OS) and endoplasmic reticulum stress (ERs) in neuronal cells. Further studies showed that the Notch1-Nrf2 signaling pathway may participate in Pin1 regulation of NTDs, as evidenced by the inhibition and overexpression of the Notch1-Nrf2 pathway. In addition, immunofluorescence (IF), co-immunoprecipitation (Co-IP), and GST pull-down experiments also showed that Pin1 interacts directly with Notch1 and Nrf2. Thus, our study suggested that the knocking down of Pin1 promotes NTD progression by inhibiting the activation of the Notch1-Nrf2 signaling pathway, and it is possible that this effect is achieved by disrupting the interaction of Pin1 with Notch1 and Nrf2, affecting their proteostasis. Our research identified that the regulation of Pin1 by retinoic acid (RA) and its involvement in the development of NTDs through the Notch1-Nrf2 axis could enhance our comprehension of the mechanism behind RA-induced brain abnormalities.


Assuntos
Peptidilprolil Isomerase de Interação com NIMA , Defeitos do Tubo Neural , Tretinoína , Animais , Feminino , Humanos , Camundongos , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Tubo Neural/metabolismo , Tubo Neural/efeitos dos fármacos , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/induzido quimicamente , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Peptidilprolil Isomerase de Interação com NIMA/genética , Estresse Oxidativo/efeitos dos fármacos , Receptor Notch1/metabolismo , Receptor Notch1/genética , Transdução de Sinais/efeitos dos fármacos , Tretinoína/metabolismo , Tretinoína/farmacologia
5.
Toxins (Basel) ; 13(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34822527

RESUMO

Fumonisin B1 (FB1) is among the most common contaminants produced by Fusarium spp. fungus from corns and animal feeds. Although FB1 has been known to cause physical or functional defects of embryos in humans and several animal species such as Syrian hamsters, rabbits, and rodents, little is known about the precise toxicity to the embryos and the underlying mechanisms have not been fully addressed. The present study aimed to investigate its developmental toxicity and potential mechanisms of action on sphingolipid metabolism in Brown Tsaiya Ducks (BTDs) embryos. We examined the effect of various FB1 dosages (0, 10, 20 and 40 µg/embryo) on BTD embryogenesis 72 h post-incubation. The sphingomyelin content of duck embryos decreased (p < 0.05) in the highest FB1-treated group (40 µg). Failure of neural tube closure was observed in treated embryos and the expression levels of a neurulation-related gene, sonic hedgehog (Shh) was abnormally decreased. The sphingolipid metabolism-related genes including N-acylsphingosine amidohydrolase 1 (ASAH1), and ceramide synthase 6 (CERS6) expressions were altered in the treated embryos compared to those in the control embryos. Apparently, FB1 have interfered sphingolipid metabolisms by inhibiting the functions of ceramide synthase and folate transporters. In conclusion, FB1-caused developmental retardation and abnormalities, such as neural tube defects in Brown Tsaiya Duck embryos, as well as are partly mediated by the disruption of sphingolipid metabolisms.


Assuntos
Patos/embriologia , Fumonisinas/efeitos adversos , Tubo Neural/efeitos dos fármacos , Esfingolipídeos/metabolismo , Animais , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/embriologia , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/embriologia
6.
Development ; 148(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34610637

RESUMO

Many developmental disorders are thought to arise from an interaction between genetic and environmental risk factors. The Hedgehog (HH) signaling pathway regulates myriad developmental processes, and pathway inhibition is associated with birth defects, including holoprosencephaly (HPE). Cannabinoids are HH pathway inhibitors, but little is known of their effects on HH-dependent processes in mammalian embryos, and their mechanism of action is unclear. We report that the psychoactive cannabinoid Δ9-tetrahydrocannabinol (THC) induces two hallmark HH loss-of-function phenotypes (HPE and ventral neural tube patterning defects) in Cdon mutant mice, which have a subthreshold deficit in HH signaling. THC therefore acts as a 'conditional teratogen', dependent on a complementary but insufficient genetic insult. In vitro findings indicate that THC is a direct inhibitor of the essential HH signal transducer smoothened. The canonical THC receptor, cannabinoid receptor-type 1, is not required for THC to inhibit HH signaling. Cannabis consumption during pregnancy may contribute to a combination of risk factors underlying specific developmental disorders. These findings therefore have significant public health relevance.


Assuntos
Padronização Corporal/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/toxicidade , Dronabinol/toxicidade , Holoprosencefalia/induzido quimicamente , Receptor Smoothened/metabolismo , Teratogênicos/toxicidade , Animais , Agonistas de Receptores de Canabinoides/farmacologia , Moléculas de Adesão Celular/genética , Células Cultivadas , Dronabinol/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Tubo Neural/metabolismo , Transdução de Sinais/efeitos dos fármacos , Teratogênicos/farmacologia
7.
Turk Neurosurg ; 31(4): 594-600, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33978217

RESUMO

AIM: To investigate the effects of pregabalin on neural tube closure, and other potential effects on other organ systems in a chick embryo model. MATERIAL AND METHODS: Fertilized chicken eggs were divided into groups, and different doses of pregabalin was administered. All embryos were harvested in the 8th day of incubation, and investigated both macroscopically and microscopically against any developmental malformations caused by Pregabalin. RESULTS: Macroscopically not any malformations were detected but macrosomia was statistically significant in medium and high dose groups. Microscopically, vertebral lamina ossification was delayed in some embryos in high dose group but not interpreted as midline closure defect and also not statistically significant. Decrease in the number of renal glomerulus and increase in the tubular damage was statistically significant in medium and high dose groups. Cardiomegaly was also found in some embryos in middle and high dose groups but not statistically significant. CONCLUSION: The use of pregabalin does not cause neural tube closure defect in the embryo unless not exceed recommended maximum dose. Causing macrosomia instead of developmental retardation by Pregabalin is in conflict with the literature. This study revealed that Pregabalin causes fetal nephrotoxicity and macrosomia. These findings indicate that the use of Pregabalin in pregnancy still needs to be accounted as suspicious.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/efeitos dos fármacos , Pregabalina/toxicidade , Teratogênese/efeitos dos fármacos , Animais , Embrião de Galinha , Galinhas/crescimento & desenvolvimento , Relação Dose-Resposta a Droga , Tubo Neural/embriologia , Tubo Neural/crescimento & desenvolvimento , Defeitos do Tubo Neural/induzido quimicamente , Pregabalina/farmacologia , Testes de Toxicidade
8.
World Neurosurg ; 150: e613-e620, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33753320

RESUMO

BACKGROUND: Neural tube defects are among the most frequent congenital abnormalities of the central nervous system. Progression of neural tube deficits is affected by hereditary predilection and environmental determinants. Pethidine (meperidine) is a fast and powerful opioid analgesic in U.S. Food and Drug Administration category C. There are reports about developmental anomalies due to this medication. The aim of this study was to investigate the effects of different doses of pethidine hydrochloride on neural tube development in a chick embryo model resembling the first month of vertebral growth in mammals. METHODS: Seventy-five specific pathogen-free eggs were incubated for 28 hours and divided into 5 groups (including the control group), each consisting of 15 eggs. Pethidine hydrochloride was administered sub-blastodermically with a Hamilton microinjector in 4 different doses. Incubation was continued until the end of the 48th hour. Subsequently, all eggs were opened, and embryos were cut from the embryonic membranes and evaluated morphologically, genetically, and histopathologically. RESULTS: Crown-rump length, somite numbers, and silver-stained nucleolar organizer region (AgNOR) number averages, and total AgNOR/nuclear area ratios decreased in a dose-dependent manner. Examination of neural tube closure revealed statistically significant differences in all experimental groups (P<0.05). Messenger RNA levels of the BRE gene were decreased in pethidine hydrochloride-exposed embryos compared with the control group. Although this downregulation was not statistically significant, this decrease was striking with a 0.422-fold change in the fifth group. CONCLUSIONS: We demonstrated that pethidine hydrochloride affects neuronal development in chicken embryos. The teratogenic mechanism of pethidine hydrochloride is unclear; therefore, further investigation is required.


Assuntos
Analgésicos Opioides/toxicidade , Meperidina/toxicidade , Tubo Neural/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Embrião de Galinha
9.
Turk Neurosurg ; 31(1): 137-141, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33491173

RESUMO

AIM: To investigate the effects of quetiapine exposure on neural tube development in early stage chicken embryos. MATERIAL AND METHODS: Eighty-four fertilised specific pathogen-free chicken eggs were divided into four equal groups (groups 1?4). Three experimental groups (groups 2, 3 and 4) and a single control group (group 1) were used. Each egg in group 2 (n=21) was injected with 20 ?L of saline after 30 hours of incubation. Eggs in groups 3 and 4 were injected with 0.02 ml of a solution containing 400 and 800 ?g of quetiapine dose, respectively. Incubation was continued until the end of 72 hours. All embryos were then removed from the eggs and histopathologically examined. RESULTS: Normal development and the closed neural tubes were shown in 18, 16, 13 and 9 embryos in groups 1 2, 3 and 9, respectively, of the 84 embryos incubated. Open neural tubes were found in one, three and five embryos in groups 2, 3 and 5, respectively. Also, developmental anomalies were found in three, four, five and seven embryos in groups 1, 2, 3 and 4, respectively. Moreover, no significant relationship between NTD and quetiapine exposure had been found. CONCLUSION: Quetiapine has no significant effect on the occurrence of neural tube defects in the chicken embryo model.


Assuntos
Antipsicóticos/administração & dosagem , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Fumarato de Quetiapina/administração & dosagem , Animais , Antipsicóticos/efeitos adversos , Embrião de Galinha , Galinhas , Desenvolvimento Embrionário/fisiologia , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/diagnóstico , Fumarato de Quetiapina/efeitos adversos
10.
Neurotoxicol Teratol ; 83: 106941, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33212164

RESUMO

Nuclear factor kappa B (NF-κB) is a heterodimer of protein subunits p65 and p50, that regulates the expression of a large number of genes related to cell growth and proliferation. The p65 subunit is activated after phosphorylation by Pim-1, while the p50 subunit is the cleaved product of its precursor molecule p105. Valproic acid (VPA), an antiepileptic drug, is a known teratogen and its exposure during pregnancy is associated with 1-2% of neural tube defects in the offspring. The current study aimed at investigating the effects of in utero VPA exposure on the key components of the NF-κB signaling pathway including p65, p50, and Pim-1 in CD-1 mouse embryos during the critical period of neural tube closure. Here we report that p65, Pim-1 and p105/p50 mRNA were significantly (p < 0.05) downregulated at 1 and 3 h following in utero exposure to a teratogenic dose (400 mg/kg) of VPA in gestational day (GD)9 exposed embryos. At GD13 heads of control, non-exencephalic and exencephalic embryos were used for analysis and we found significant upregulation of p65 protein expression in non-exencephalic GD13 heads while p50 protein levels were significantly downregulated in both non-exencephalic and exencephalic groups. On the other hand, p65 and p50 protein levels remained unchanged in the nuclear extracts of the VPA-exposed non-exencephalic and exencephalic GD13 embryo heads. The reported results suggest that VPA exposure perturbates p65, p105/p50, Pim-1 transcript and p65/p50 protein levels in mouse embryos.


Assuntos
NF-kappa B/metabolismo , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Ácido Valproico/toxicidade , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/toxicidade , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Troca Materno-Fetal , Camundongos , NF-kappa B/genética , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Tubo Neural/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/metabolismo , Neurotoxinas/administração & dosagem , Neurotoxinas/toxicidade , Neurulação/efeitos dos fármacos , Neurulação/fisiologia , Gravidez , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Ácido Valproico/administração & dosagem
11.
Alcohol Clin Exp Res ; 44(8): 1540-1550, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32557641

RESUMO

BACKGROUND: Early gestational alcohol exposure is associated with severe craniofacial and CNS dysmorphologies and behavioral abnormalities during adolescence and adulthood. Alcohol exposure during the formation of the neural tube (gestational day [GD] 8 to 10 in mice; equivalent to4th week of human pregnancy) disrupts development of ventral midline brain structures such as the pituitary, septum, and ventricles. This study identifies transcriptomic changes in the rostroventral neural tube (RVNT), the region of the neural tube that gives rise to the midline structures sensitive to alcohol exposure during neurulation. METHODS: Female C57BL/6J mice were administered 2 doses of alcohol (2.9 g/kg) or vehicle 4 hours apart on GD 9.0. The RVNTs of embryos were collected 6 or 24 hours after the first dose and processed for RNA-seq. RESULTS: Six hours following GD 9.0 alcohol exposure (GD 9.25), over 2,300 genes in the RVNT were determined to be differentially regulated by alcohol. Enrichment analysis determined that PAE affected pathways related to cell proliferation, p53 signaling, ribosome biogenesis, and immune activation. In addition, over 100 genes involved in primary cilia formation and function and regulation of morphogenic pathways were altered 6 hours after alcohol exposure. The changes to gene expression were largely transient, as only 91 genes identified as differentially regulated by prenatal alcohol at GD 10 (24 hours postexposure). Functionally, the differentially regulated genes at GD 10 were related to organogenesis and cell migration. CONCLUSIONS: These data give a comprehensive view of the changing landscape of the embryonic transcriptome networks in regions of the neural tube that give rise to brain structures impacted by a neurulation-stage alcohol exposure. Identification of gene networks dysregulated by alcohol will help elucidate the pathogenic mechanisms of alcohol's actions.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Embrião de Mamíferos/efeitos dos fármacos , Etanol/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Tubo Neural/efeitos dos fármacos , Neurulação/efeitos dos fármacos , Animais , Proliferação de Células/genética , Cílios/genética , Embrião de Mamíferos/metabolismo , Feminino , Perfilação da Expressão Gênica , Camundongos , Tubo Neural/metabolismo , Neurulação/genética , Biogênese de Organelas , Gravidez , RNA-Seq , Ribossomos/genética , Proteína Supressora de Tumor p53
12.
Turk Neurosurg ; 30(4): 583-587, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32530477

RESUMO

AIM: To investigate the effects of tartrazine exposure on neural tube development, in early stage chicken embryos. MATERIAL AND METHODS: A total of 120 fertilized specific pathogen-free chicken eggs were divided into 4 equal groups (groups 1?4). After 30 hours of incubation, the eggs, except for the Group 1 (control group), were opened under 4X optical magnification. Group 2 was administered physiological saline. Group 3 was administered a middle dose of tartrazin (4.5 mg/kg) at a volume of 20 µL by the in ovo method, and group 4 was administered a high dose of tartrazine (7.5 mg/kg) using the same process. Incubation was continued until the end of the 72nd hour; all embryos were then removed from the eggs and histopathologically examined. RESULTS: Of the 120 embryos incubated, normal development and the closed neural tubes were shown in all embryos in group 1; 23 in group 2; 19 in group 3 and; only 9 in group 4. Open neural tubes were found in; 4 embryos in group 2; 5 embryos in group 3 and; 13 embryos in group 4. The neural tube closure defect was found to be significantly higher in group 4 compared to the other groups (p < 0.01). CONCLUSION: Based on our data, tartrazine, as one of the widely used food coloring agent, was seen to cause a neural tube defect in the chicken embryo model.


Assuntos
Corantes de Alimentos/toxicidade , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/patologia , Tubo Neural/efeitos dos fármacos , Tartrazina/toxicidade , Animais , Embrião de Galinha , Galinhas , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/fisiologia , Tubo Neural/patologia
13.
Cell Mol Neurobiol ; 40(3): 383-393, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31555941

RESUMO

Neural crest cells (NCCs) comprise a population of multipotent progenitors and stem cells at the origin of the peripheral nervous system (PNS) and melanocytes of skin, which are profoundly influenced by microenvironmental factors, among which is basic fibroblast growth factor 2 (FGF2). In this work, we further investigated the role of this growth factor in quail trunk NC morphogenesis and demonstrated its huge effect in NCC growth mainly by stimulating cell proliferation but also reducing cell death, despite that NCC migration from the neural tube explant was not affected. Moreover, following FGF2 treatment, reduced expression of the early NC markers Sox10 and FoxD3 and improved proliferation of HNK1-positive NCC were observed. Since these markers are involved in the regulation of glial and melanocytic fate of NC, the effect of FGF2 on NCC differentiation was investigated. Therefore, in the presence of FGF2, increased proportions of NCCs positives to the melanoblast marker Mitf as well as NCCs double stained to Mitf and BrdU were recorded. In addition, treatment with FGF2, followed by differentiation medium, resulted in increased expression of melanin and improved proportion of melanin-pigmented melanocytes without alteration in the glial marker Schwann myelin protein (SMP). Taken together, these data further reveal the important role of FGF2 in NCC proliferation, survival, and differentiation, particularly in melanocyte development. This is the first demonstration of FGF2 effects in melanocyte commitment of NC and in the proliferation of Mitf-positive melanoblasts. Elucidating the differentiation process of embryonic NCCs brings us a step closer to understanding the development of the PNS and then undertaking the search for advanced technologies to prevent, or treat, injuries caused by NC-related disorders, also known as neurocristopathies.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Melanócitos/efeitos dos fármacos , Crista Neural/efeitos dos fármacos , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Células-Tronco Embrionárias/fisiologia , Melaninas/metabolismo , Melanócitos/fisiologia , Crista Neural/citologia , Tubo Neural/citologia , Tubo Neural/efeitos dos fármacos , Nervos Periféricos/citologia , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/fisiologia , Codorniz/embriologia , Tronco
14.
World Neurosurg ; 134: e822-e825, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31715407

RESUMO

OBJECTIVE: Erythrosine (E127), a synthetic food dye containing iodine and sodium, has often been used inside packaged foods and beverages in Turkey and many other countries. We evaluated the effects of erythrosine on neural tube development in early-stage chicken embryos. METHODS: The study included 4 groups, with a total of 80 embryos: a control group, a normal saline group, a half-dose group, and a high-dose group. After 30 hours of incubation, saline and erythrosine solution was injected under the embryonic discs. At the end of 72 hours, the embryos were excised and evaluated macroscopically and histopathologically. RESULTS: Neural tube defects were detected in the erythrosine-administered groups with statistically significant differences. In contrast, the embryos in the control and saline groups displayed normal development. CONCLUSIONS: Erythrosine increased the risk of neural tube defects in early-stage chicken embryos, even at half of the approved dose.


Assuntos
Eritrosina/farmacologia , Corantes Fluorescentes/farmacologia , Defeitos do Tubo Neural/embriologia , Tubo Neural/efeitos dos fármacos , Anormalidades Induzidas por Medicamentos/embriologia , Anormalidades Induzidas por Medicamentos/etiologia , Animais , Embrião de Galinha , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/embriologia , Defeitos do Tubo Neural/induzido quimicamente
15.
Turk Neurosurg ; 29(6): 851-855, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31192442

RESUMO

AIM: To investigate the effects of Phenyramidol (Phe) on neural development in an early chicken embryo model. MATERIAL AND METHODS: Sixty fertile non-pathogenic Super Nick eggs were incubated for 24 hours (h) and divided into four groups of 15 eggs each. Phe was administrated through the sub-blastoderm, and the eggs were incubated for another 24 h. All eggs were opened after 48 h of incubation, and the embryos were evaluated morphologically and histopathologically. RESULTS: In Group 1 (control group), none exhibited neural tube defects (NTDs) (0%), 1 (6.6%) was undeveloped; in Group 2 (low dosages), 1 did not develop (6.6%); in Group 3 (normal dosages), 2 (13.4%) had NTDs, 1 (6.6%) was undeveloped; in Group 4 (high dosages), 5 (33.3%) had NTDs, 2 (13.3%) were undeveloped. CONCLUSION: In light of the results, it was determined that the use of increasing doses of Phe led to defects in midline closure in early chicken embryos. This is the first report in the literature on Phe used in an early chicken embryo model.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Relaxantes Musculares Centrais/toxicidade , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Piridinas/toxicidade , Animais , Embrião de Galinha , Galinhas , Desenvolvimento Embrionário/fisiologia , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/patologia
16.
Acta Neurobiol Exp (Wars) ; 79(1): 92-100, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31038488

RESUMO

In a previous study, methylenedioxypyrovalerone (MDPV), a designer drug of the cathinone family, caused selective enhancement of Caspase3 immunoreactive (Casp3+) apoptotic cells in the nucleus accumbens (NAc) of 7­day­old mice. To further elaborate on the mechanism underlying MDPV­elicited apoptosis, here, we investigated the appearance of Casp3+ cells in developing neural tube explants of E12.5 mice, following MDPV treatment in vitro. Apoptotic cells appeared in large number in the pallium as radial progenitor cells and multipolar neurons, and in the subpallium including the future NAc, both in control and MDPV treated specimens. MDPV did not cause gross morphological changes in the neural tube or in the abundance of Casp3+ cells, based on a visual impression, though quantification was not attempted. We also studied the changes in NMDA receptor (NMDAR) protein subunits NR1 and NR2B in the NAc of 7­day­old MDPV treated and control mice, using western blotting of tissue obtained by selective dissection. In MDPV treated animals, expression of NR2B was lower than in the control animals, whereas expression of NR1 did not differ significantly from controls. The findings indicate that, during early postembryonic development, downregulation of the NR2B receptor subunit (at this time predominant in the NMDAR) is accompanied by a decreased viability of neurons. Decreased viability is expressed, in this case, as enhanced susceptibility to stimulation by MDPV - essentially a robust dopaminergic agent, potently affecting the neurons of the NAc. The findings are likely relevant to dopaminergic/NMDAR interactions and a potential pro­survival role of the NR2B subunit in critical phases of neural development.


Assuntos
Apoptose/efeitos dos fármacos , Benzodioxóis/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Núcleo Accumbens/citologia , Pirrolidinas/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Caspase 3/metabolismo , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos BALB C , Tubo Neural/citologia , Tubo Neural/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Catinona Sintética
17.
Ecotoxicol Environ Saf ; 180: 192-201, 2019 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-31085430

RESUMO

As a main marine phycotoxin, okadaic acid (OA) is mainly responsible for diarrheic shellfish poisoning (DSP), through specifically inhibiting phosphatase (PP1 and PP2A). It has been shown that isotope labelled-OA could cross the placental barrier in mice. However, it remains obscure how OA exposure could affect the formation of neural crest cells (NCCs), especially cranial NCCs in early embryo development. Here, we explored the effects of OA exposure on the generation of neural crest cells during embryonic development using the classic chick embryo model. We found that OA exposure at 100 nM (80.5 µg/L) could cause craniofacial bone defects in the developing chick embryo and delay the development of early chick embryos. Immunofluorescent staining of HNK-1, Pax7, and Ap-2α demonstrated that cranial NCC generation was inhibited by OA exposure. Double immunofluorescent staining with Ap-2α/PHIS3 or Pax7/c-Caspase3 manifested that both NCC proliferation and apoptosis were restrained by OA exposure. Furthermore, the expression of Msx1 and BMP4 were down-regulated in the developing chick embryonic neural tubes, which could contribute the inhibitive production of NCCs. We also discovered that expression of EMT-related adhesion molecules, such as Cadherin 6B (Cad6B) and E-cadherin, was altered following OA exposure. In sum, OA exposure negatively affected the development of embryonic neural crest cells, which in turn might result in cranial bone malformation.


Assuntos
Inibidores Enzimáticos/toxicidade , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Crista Neural/efeitos dos fármacos , Ácido Okadáico/toxicidade , Animais , Apoptose/efeitos dos fármacos , Caderinas/metabolismo , Embrião de Galinha , Regulação para Baixo , Desenvolvimento Embrionário/efeitos dos fármacos , Crista Neural/citologia , Crista Neural/embriologia , Tubo Neural/efeitos dos fármacos , Tubo Neural/metabolismo , Crânio/anormalidades
18.
Int J Dev Biol ; 63(1-2): 37-43, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30919914

RESUMO

The receptors of gamma-aminobutyric acid (GABA), which is a well-known neurotransmitter, are expressed in the anterior-to-mid neural tube at an early stage of Xenopus development, but there has been no report on the role of GABA in the presumptive central nervous system. Therefore, we tried to reveal the function of GABA for Xenopus early embryogenesis. We first confirmed that the region expressing a gene encoding glutamate decarboxylase 1 (gad1), which is an enzyme that catalyzes the decarboxylation of L-glutamate to GABA, overlapped with that of several genes encoding GABA receptors (gabr) in the neural tube. Metabolome analysis of culture medium of dorsal tail-bud explants containing the neural tube region of tail-bud stage embryos also revealed that GABA was expressed at this stage. Then, we examined the treatment of pentylenetetrazole (PTZ) and picrotoxin (PTX), which are known as inhibitors of GABA receptors (GABA-R), on the early stages of Xenopus embryogenesis, and found that axis elongation in the tail-bud was inhibited by both treatments, and these phenotypic effects were rescued by co-treatment with GABA. Moreover, our spatial- and temporal-specific inhibitor treatments revealed that the gabr- and gad1-overlapped region, which presents at the anterior-to-mid neural tube during the tail-bud stages, was much more sensitive to PTZ and thus caused severe inhibition of axis elongation. Taken together, our results indicate that the small ligand molecule GABA functions as a regulator to induce the axis elongation event in the tail-bud during early embryogenesis via direct stimulation of the neural tube and indirect stimulation of the surrounding area.


Assuntos
Embrião não Mamífero/citologia , Morfogênese , Tubo Neural/embriologia , Cauda/embriologia , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Ácido gama-Aminobutírico/farmacologia , Animais , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , GABAérgicos/farmacologia , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Metaboloma , Tubo Neural/efeitos dos fármacos , Tubo Neural/metabolismo , Cauda/efeitos dos fármacos , Cauda/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo
19.
Turk Neurosurg ; 29(1): 106-109, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29806076

RESUMO

AIM: To investigate the impact of rizatriptan on neural tube development using early chick embryos as a model organism. MATERIAL AND METHODS: A total of 36 pathogen-free Leghorn chicken eggs were selected and categorized in three groups: sham, therapeutic, and supra-therapeutic. After 24 hours, the eggs were opened and injected with sterile drugs, and then reclosed using plastic tape. After a period of 72 hours, the eggs were opened and assessed using the Hamburger-Hamilton chick embryology classification method. TUNEL staining was used to identify apoptosis, and hematoxylin-eosin staining was used to investigate neural tube closure. RESULTS: Treatment with rizatriptan significantly slowed down neural tube development. The supra-therapeutic group showed neural tube closure defects. CONCLUSION: Rizatriptan had a negative effect on neural tube closure. Further research is needed to identify a safe and effective drug for treating migraines during pregnancy.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Defeitos do Tubo Neural/induzido quimicamente , Tubo Neural/efeitos dos fármacos , Agonistas do Receptor de Serotonina/toxicidade , Triazóis/toxicidade , Triptaminas/toxicidade , Animais , Embrião de Galinha , Galinhas , Tubo Neural/embriologia
20.
Folia Morphol (Warsz) ; 78(2): 307-313, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30178461

RESUMO

BACKGROUND: Neural tube defects are congenital malformations of the central nervous system. Genetic predisposition and some environmental factors play an important role in the development of neural tube defects. This study aimed to investigate the effects of diclofenac sodium on the neural tube development in a chick embryo model that corresponds to the first month of vertebral deve- lopment in mammals. MATERIALS AND METHODS: Seventy-five fertile, specific pathogen-free eggs were incubated for 28 h and were divided into five groups of 15 eggs each. Diclofenac sodium was administered via the sub-blastodermic route at this stage. Incubation was continued till the end of the 48th h. All eggs were then opened and embryos were dissected from embryonic membranes and evaluated morphologically and histopathologically. RESULTS: It was determined that the use of increasing doses of diclofenac sodium led to defects of midline closure in early chicken embryos. There were statistically significant differences in neural tube positions (open or close) among the groups. In addition; crown-rump length, somite number were significantly decreased in high dose experimental groups compared with control group. CONCLUSIONS: This study showed that development of neurons is affected in chi- cken embryos after administration of diclofenac sodium. The exact teratogenic mechanism of diclofenac sodium is not clear; therefore it should be investigated.


Assuntos
Diclofenaco/efeitos adversos , Tubo Neural/embriologia , Animais , Embrião de Galinha , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/efeitos dos fármacos , Tubo Neural/patologia , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/patologia , Estatística como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA