Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 964
Filtrar
1.
Toxicol Lett ; 359: 55-64, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35122893

RESUMO

Methylmercury (MeHg) is a bio-accumulative global environmental contaminant present in fish and seafood. MeHg accumulates in the aquatic environment and eventually reaches the human system via the food chain by bio-magnification. The central nervous system is the primary target of toxicity and is particularly vulnerable during development. It is well documented that developmental MeHg exposure can lead to neurological alterations, including cognitive and motor dysfunction. Apoptosis is a primary characteristic of MeHg-induced neurotoxicity, and may be regulated by autophagic activity. However, mechanisms mediating the interaction between apoptosis and autophagy remains to be explored. Autophagy is an adaptive response under stressful conditions, and the basal level of autophagy ensures the physiological turnover of old and damaged organelles. Autophagy can regulate cell fate through different crosstalk signaling pathways. A complex interplay between autophagy and apoptosis determines the degree of apoptosis and the progression of MeHg-induced neurotoxicity as demonstrated by pre-clinical models and clinical trials. This review summarizes recent advances in the roles of autophagy and apoptosis in MeHg neurotoxicity and thoroughly explores the relationship between them. The autophagic pathway may be a potential therapeutic target in MeHg neurotoxicity through modulation of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Compostos de Metilmercúrio/metabolismo , Compostos de Metilmercúrio/toxicidade , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Redes e Vias Metabólicas
2.
Neuropharmacology ; 206: 108939, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-34986414

RESUMO

Methotrexate (MTX) is widely employed for children with cancer, but is also associated with persistent cognitive deficits among survivors. The present study investigated the mechanisms behind long-term cognitive dysfunction after juvenile animals are treated with MTX. Male and female Long-Evans rats were treated with a combination of 6 systemic doses (0.5 mg/kg/dose intraperitoneally) and 4 intrathecal doses (1 mg/kg) beginning at post-natal age 3 weeks, a schedule designed to mimic repeated exposure given to children with leukemia. Behavioral testing was conducted at 60-61 weeks of age, followed by analysis of brain histolopathology. This MTX regimen had no acute toxicity and no effect on growth. The spatial memory and visual memory deficits observed at 13 and 17 weeks of age persisted 1 year after MTX exposure in both females and males. Significantly decreased cell proliferation and increased hippocampal microglial activation were observed in MTX-treated females when compared to the controls, with a similar trend in the male groups. In addition, MTX treatment significantly increased the number of TUNEL positive cells in the periventricular area. Our study demonstrates that a clinically relevant regimen of systemic and intrathecal MTX induces persistent deficits in cognition, lasting approximately 1 year after the last injection. The mechanisms behind MTX-induced deficits are likely multifactorial, including suppression of neurogenesis, microglial activation, and increased brain cell apoptosis. Our study suggests female and male animals differ in susceptibility to MTX-induced neurotoxicity and provides insights for developing therapeutic approaches to prevent treatment related cognitive impairment among children with ALL.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/fisiopatologia , Metotrexato/farmacologia , Síndromes Neurotóxicas/fisiopatologia , Fatores Etários , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Sobreviventes de Câncer , Modelos Animais de Doenças , Feminino , Masculino , Metotrexato/administração & dosagem , Microglia/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Ratos , Ratos Long-Evans , Fatores Sexuais , Fatores de Tempo
3.
Hum Exp Toxicol ; 41: 9603271211061502, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35023765

RESUMO

Ethylene glycol (EG), in addition to its neurotoxic and nephrotoxic effects, evokes oxidative stress. The aim of this study was to assess the influence of the ethylene glycol on the biochemical indicators and oxidoreductive balance of patients treated for acute poisoning. The total study group consisted of 56 persons including 26 alcoholics who took EG as a substitute for ethyl alcohol in the course of alcohol dependence syndrome and 30 controls. Severity of poisoning, results of acid-base parameters, biochemical, and toxicological tests as well as biomarkers of the oxidative stress in blood were analyzed during the patients' hospitalization. The key issue was to assess the oxidative stress and biochemical disturbances caused by EG and the type of treatment applied in the course of poisoning. Significant changes in some parameters were found both at time of diagnosis and after treatment initiation (ethanol as an antidote and hemodialysis). The most important differences included the activity of hepatic parameters (aspartate aminotransferase, AST) and oxidative stress markers like catalase (CAT); correlation of the lipid peroxidation products level (TBARS) with urea concentration has been shown. On the last day of the hospitalization, in some cases, the mutual correlation between the evaluated markers were observed, for example, between alanine transaminase (ALT) and glutathione reductase (GR), and urea concentration and glutathione level (GSH/GSSG). The concentration of ions (H+) had a major impact on the oxidoreductive balance, correlating with the elevated GR and GSH/GSSG levels.


Assuntos
Alcoolismo/complicações , Alcoolismo/fisiopatologia , Etanol/envenenamento , Etilenoglicóis/envenenamento , Fomepizol/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/fisiopatologia , Adulto , Antídotos/uso terapêutico , Biomarcadores/sangue , Etanol/sangue , Etilenoglicóis/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Neurotóxicas/etiologia , Estresse Oxidativo/efeitos dos fármacos
4.
Toxicology ; 465: 153030, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34774978

RESUMO

Over the years, the advancement of radio diagnostic imaging tools and techniques has radically improved the diagnosis of different pathophysiological conditions, accompanied by increased exposure to low-dose ionizing radiation. Though the consequences of high dose radiation exposure on humans are very well comprehended, the more publicly relevant effects of low dose radiation (LDR) (≤100 mGy) exposure on the biological system remain ambiguous. The central nervous system, predominantly the developing brain with more neuronal precursor cells, is exceptionally radiosensitive and thus more liable to neurological insult even at low doses, as shown through several rodent studies. Further molecular studies have unraveled the various inflammatory and signaling mechanisms involved in cellular damage and repair that drive these physiological alterations that lead to functional alterations. Interestingly, few studies also claim that LDR exerts therapeutic effects on the brain by initiating an adaptive response. The present review summarizes the current understanding of the effects of low dose radiation at functional, cellular, and molecular levels and the various risks and benefits associated with it based on the evidence available from in vitro, in vivo, and clinical studies. Although the consensus indicates minimum consequences, the overall evidence suggests that LDR can bring about considerable neurological effects in the exposed individual, and hence a re-evaluation of the LDR usage levels and frequency of exposure is required.


Assuntos
Comportamento Animal/efeitos da radiação , Encéfalo/efeitos da radiação , Síndromes Neurotóxicas/etiologia , Doses de Radiação , Exposição à Radiação/efeitos adversos , Lesões por Radiação/etiologia , Radiação Ionizante , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Relação Dose-Resposta à Radiação , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/fisiopatologia , Lesões por Radiação/metabolismo , Lesões por Radiação/patologia , Lesões por Radiação/fisiopatologia , Medição de Risco , Fatores de Risco , Transdução de Sinais/efeitos da radiação
5.
Toxicol Lett ; 354: 44-55, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34757178

RESUMO

Disruption of the thyroid hormone system during development can impair brain development and cause irreversible damage. Some thyroid hormone system disruptors act by inhibiting the thyroperoxidase (TPO) enzyme, which is key to thyroid hormone synthesis. For the potent TPO-inhibiting drug propylthiouracil (PTU) this has been shown to result in thyroid hormone system disruption and altered brain development in animal studies. However, an outstanding question is which chemicals beside PTU can cause similar effects on brain development and to what degree thyroid hormone insufficiency must be induced to be able to measure adverse effects in rats and their offspring. To start answering these questions, we performed a perinatal exposure study in pregnant rats with two TPO-inhibitors: the drug methimazole (MMI) and the triazole herbicide amitrole. The study involved maternal exposure from gestational day 7 through to postnatal day 22, to MMI (8 and 16 mg/kg body weight/day) or amitrole (25 and 50 mg/kg body weight/day). Both MMI and amitrole reduced serum T4 concentrations in a dose-dependent manner in dams and offspring, with a strong activation of the hypothalamic-pituitary-thyroid axis. This reduction in serum T4 led to decreased thyroid hormone-mediated gene expression in the offspring's brains and caused adverse effects on brain function, seen as hyperactivity and decreased habituation in preweaning pups. These dose-dependent effects induced by MMI and amitrole are largely the same as those observed with PTU. This demonstrates that potent TPO-inhibitors can induce effects on brain development in rats and that these effects are driven by T4 deficiency. This knowledge will aid the identification of TPO-inhibiting thyroid hormone system disruptors in a regulatory context and can serve as a starting point in search of more sensitive markers of developmental thyroid hormone system disruption.


Assuntos
Amitrol (Herbicida)/toxicidade , Antitireóideos/toxicidade , Inibidores Enzimáticos/toxicidade , Metimazol/toxicidade , Atividade Motora/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Glândula Tireoide/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Exposição Materna/efeitos adversos , Síndromes Neurotóxicas/fisiopatologia , Gravidez , Ratos , Transdução de Sinais/efeitos dos fármacos , Testes de Função Tireóidea
6.
Toxicology ; 465: 153052, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34838597

RESUMO

Bisphenol-A (BPA), an environmental endocrine disruptor, is toxic to the central nervous system. Although recent studies have shown BPA-induced neurotoxicity, it is far from clear what precisely epigenetic mechanisms are involved in BPA-induced cognitive deficits. In this study, pheochromocytoma (PC12) cells were treated with BPA at 1 µM for 36 h in vitro. In vivo, C57BL/6 mice were administered to BPA at a dose of 1 mg/kg/day for 10 weeks. The results showed that 1 µM BPA exposure for 36 h impaired neurite outgrowth of PC12 cells through decreasing the primary and secondary branches. Besides, BPA exposure decreased the level of Ac-H3K9 (histone H3 Lys9 acetylation) by upregulating the expression of HDAC2 (histone deacetylases 2) in PC12 cells. Furthermore, treatment of both TSA (Trichostatin A, inhibitor of the histone deacetylase) and shHDAC2 plasmid (HDAC2 knockdown construct) resulted in amelioration neurite outgrowth deficits induced by BPA. In addition, it was shown that repression of HDAC2 could markedly rescue the spine density impairment in the hippocampus and prevent the cognitive impairment caused by BPA exposure in mice. Collectively, HDAC2 plays an essential role in BPA-induced neurotoxicity, which provides a potential molecular target for medical intervention.


Assuntos
Compostos Benzidrílicos/toxicidade , Espinhas Dendríticas/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Hipocampo/efeitos dos fármacos , Histona Desacetilase 2/metabolismo , Neuritos/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Fenóis/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Cognição/efeitos dos fármacos , Espinhas Dendríticas/enzimologia , Espinhas Dendríticas/patologia , Feminino , Hipocampo/enzimologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Histona Desacetilase 2/genética , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Neuritos/enzimologia , Neuritos/patologia , Crescimento Neuronal/efeitos dos fármacos , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/fisiopatologia , Células PC12 , Ratos , Regulação para Cima
7.
Pharmacol Res ; 174: 105921, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34601079

RESUMO

Neurotoxicity is a frequent side effect of cisplatin (CisPt)-based anticancer therapy whose pathophysiology is largely vague. Here, we exploited C. elegans as a 3R-compliant in vivo model to elucidate molecular mechanisms contributing to CisPt-induced neuronal dysfunction. To this end, we monitored the impact of CisPt on various sensory functions as well as pharyngeal neurotransmission by recording electropharyngeograms (EPGs). CisPt neither affected food and odor sensation nor mechano-sensation, which involve dopaminergic and glutaminergic neurotransmission. However, CisPt reduced serotonin-regulated pharyngeal pumping activity independent of changes in the morphology of related neurons. CisPt-mediated alterations in EPGs were fully rescued by addition of serotonin (5-HT) (≤ 2 mM). Moreover, the CisPt-induced pharyngeal injury was prevented by co-incubation with the clinically approved serotonin re-uptake inhibitory drug duloxetine. A protective effect of 5-HT was also observed with respect to CisPt-mediated impairment of another 5-HT-dependent process, the egg laying activity. Importantly, CisPt-induced apoptosis in the gonad and learning disability were not influenced by 5-HT. Using different C. elegans mutants we found that CisPt-mediated (neuro)toxicity is independent of serotonin biosynthesis and re-uptake and likely involves serotonin-receptor subtype 7 (SER-7)-related functions. In conclusion, by measuring EPGs as a surrogate parameter of neuronal dysfunction, we provide first evidence that CisPt-induced neurotoxicity in C. elegans involves 5-HT-dependent neurotransmission and SER-7-mediated signaling mechanisms and can be prevented by the clinically approved antidepressant duloxetine. The data highlight the particular suitability of C. elegans as a 3R-conform in vivo model in molecular (neuro)toxicology and, moreover, for the pre-clinical identification of neuroprotective candidate drugs.


Assuntos
Antineoplásicos/toxicidade , Caenorhabditis elegans/efeitos dos fármacos , Cisplatino/toxicidade , Modelos Animais de Doenças , Síndromes Neurotóxicas/metabolismo , Serotonina/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Cloridrato de Duloxetina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/fisiopatologia , Faringe/efeitos dos fármacos , Faringe/fisiologia , Inibidores da Recaptação de Serotonina e Norepinefrina/farmacologia , Transmissão Sináptica/efeitos dos fármacos
8.
Regul Toxicol Pharmacol ; 127: 105064, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34656748

RESUMO

Over the last decade, immunotherapy has established itself as an important novel approach in the treatment of cancer, resulting in a growing importance in oncology. Engineered T cell therapies, namely chimeric antigen receptor (CAR) T cells and T cell receptor (TCR) T cell therapies, are platform technologies that have enabled the development of products with remarkable efficacy in several hematological malignancies and are thus the focus of intense research and development activity. While engineered T cell therapies offer promise in addressing currently intractable cancers, they also present unique challenges, including their nonclinical safety assessment. A workshop organized by HESI and the US Food and Drug Administration (FDA) was held to provide an interdisciplinary forum for representatives of industry, academia and regulatory authorities to share information and debate on current practices for the nonclinical safety evaluation of engineered T cell therapies. This manuscript leverages what was discussed at this workshop to provide an overview of the current important nonclinical safety assessment considerations for the development of these therapeutic modalities (cytokine release syndrome, neurotoxicity, on-target/off-tumor toxicities, off-target effects, gene editing or vector integration-associated genomic injury). The manuscript also discusses approaches used for hazard identification or risk assessment and provides a regulatory perspective on such aspects.


Assuntos
Engenharia Celular/métodos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Linfócitos T/imunologia , Síndrome da Liberação de Citocina/fisiopatologia , Edição de Genes , Imunoterapia Adotiva/efeitos adversos , Síndromes Neurotóxicas/fisiopatologia , Receptores de Antígenos de Linfócitos T/fisiologia , Medição de Risco
9.
J Cell Biochem ; 122(11): 1594-1608, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34494298

RESUMO

Soluble oligomers of amyloid-ß (Aß) are recognized as key pernicious species in Alzheimer's disease (AD) that cause synaptic dysfunction and memory impairments. Numerous studies have identified various types of Aß oligomers having heterogeneous peptide length, size distribution, structure, appearance, and toxicity. Here, we review the characteristics of soluble Aß oligomers based on their morphology, size, and structural reactivity toward the conformation-specific antibodies and then describe their formation, localization, and cellular effects in AD brains, in vivo and in vitro. We also summarize the mechanistic pathways by which these soluble Aß oligomers cause proteasomal impairment, calcium dyshomeostasis, inhibition of long-term potentiation, apoptosis, mitochondrial damage, and cognitive decline. These cellular events include three distinct molecular mechanisms: (i) high-affinity binding with the receptors for Aß oligomers such as N-methyl- d-aspartate receptors, cellular prion protein, nerve growth factor, insulin receptors, and frizzled receptors; (ii) the interaction of Aß oligomers with the lipid membranes; (iii) intraneuronal accumulation of Aß by α7-nicotinic acetylcholine receptors, apolipoprotein E, and receptor for advanced glycation end products. These studies indicate that there is a pressing need to carefully examine the role of size, appearance, and the conformation of oligomers in identifying the specific mechanism of neurotoxicity that may uncover potential targets for designing AD therapeutics.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Imunoterapia/métodos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/química , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Humanos , Imagem Molecular , Proteínas do Tecido Nervoso/metabolismo , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/fisiopatologia , Príons/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo
10.
Immunotherapy ; 13(15): 1261-1269, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34558978

RESUMO

Aims: Neurotoxicity (NT) is a common complication of chimeric antigen receptor (CAR) T-cell therapy. Data on early clinical identifiers for impending severe NT are lacking. Methods: The authors performed a retrospective study on 26 adult relapsed/refractory diffuse large B cell lymphoma patients treated with commercial CAR T-cell therapy (December 2017 - September 2018). Results: NT of any grade and severe NT occurred in 88 and 31% of patients, respectively. Dysgraphia (p < 0.01), disorientation (p = 0.01) and inattention (p = 0.018) were associated with severe NT, with positive predictive values of 100, 87.5 and 87.5%, respectively. Dysnomia was not associated with severe NT. Conclusion: In the authors' limited cohort, the dysgraphia, disorientation and inattention components of the CAR T-cell therapy-associated toxicity 10 scoring system were significantly associated with and predictive of impending severe NT.


Lay abstract Neurotoxicity (NT) is a common complication of chimeric antigen receptor (CAR) T-cell therapy. Information on early signs and symptoms of impending severe NT is lacking. The authors studied 26 adult patients with relapsed/refractory diffuse large B cell lymphoma who received commercial CAR T-cell therapy for the development of NT. The authors found that NT was common, with 31% of patients experiencing severe NT. In this relatively small patient population, the authors found that impaired writing, disorientation and poor attention, which are components of the CAR T-cell therapy-associated toxicity 10 scoring system, were significantly associated with and predictive of impending severe NT.


Assuntos
Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Síndromes Neurotóxicas/imunologia , Síndromes Neurotóxicas/fisiopatologia , Receptores de Antígenos Quiméricos/imunologia , Idoso , Antígenos CD19/imunologia , Humanos , Linfoma Difuso de Grandes Células B/imunologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
11.
Toxicology ; 462: 152960, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34555453

RESUMO

Picolinic acid (PIC) is a byproduct of tryptophan catabolism through the kynurenine pathway, with anabolic effects on bone in vivo and in vitro. Hence, PIC has been nominated as a possible candidate to treat and/or prevent osteoporosis. However, the effective dose and toxicity of PIC are not known yet. To test the effect of escalating and very high doses of oral PIC, male Sprague-Dawley rats were gavaged PIC: Group 1 (n = 3) received incremental doses of 125, 250 and 500 mg/kg/day PIC on days 1, 3 and 5. Group 2 (n = 3) received 500 mg/kg BID (8 h apart; i.e. 1000 mg/kg/day) PIC on Day 1. Group 3 (n = 3) received 125 mg/kg/day PIC for seven consecutive days. Group 4 (n = 3) received 250 mg/kg/day PIC for seven consecutive days. Groups 1, 3 and 4 rats were euthanized on Day 8. Group 5 (n = 6) received 500 mg/kg/day PIC for two consecutive days and then once a week dose (Days 9, 16 and 23) of 500 mg/kg/dose PIC, until euthanasia (Day 30). Blood and cerebrospinal fluid (CSF) were sampled at euthanasia, and tissues showing abnormalities at necropsy underwent histopathology evaluation. All rats displayed some degree of mild hypercalcemia and hyperkalemia. Rats receiving high doses (500 or 1000 mg/kg/day) of PIC died or were euthanized on humane grounds within the first week after showing clinical neurological signs, with animals later revealed to have brain necrosis and hemorrhage at histopathology. Rats receiving lower doses (125 or 250 mg/kg/day) of PIC completed treatment course without apparent clinical adverse events. In summary, very high doses of PIC (≥500 mg/kg/day) were vascular-neurotoxic. Possible future experiments must consider significantly lower doses.


Assuntos
Hiperpotassemia/induzido quimicamente , Síndromes Neurotóxicas/etiologia , Ácidos Picolínicos/toxicidade , Animais , Relação Dose-Resposta a Droga , Hipercalcemia/induzido quimicamente , Masculino , Síndromes Neurotóxicas/fisiopatologia , Ácidos Picolínicos/administração & dosagem , Ratos , Ratos Sprague-Dawley
12.
Toxicology ; 462: 152959, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34560124

RESUMO

The widespread use of lead (Pb) has caused global contamination, inevitable human exposure, and public health problems. Pb neurotoxicity has been linked to various human diseases, but its associated mechanism causing neurotoxicity is unknown. Drosophila melanogaster as a model organism has been used to study the mechanism involved in Pb-caused neurotoxicity and the potential role of antioxidants in ameliorating its harmful effects. The larval feeding technique was adopted to administer different concentrations of Pb (0.2-0.8 mM) to Oregon-R (ORR), superoxide dismutase (Sod), or catalase (Cat) overexpressing, and Sod or Cat knockdown flies to analyse Pb load, oxidative stress components, DNA damage, apoptosis and vacuolation in the brain. The results revealed that Pb accumulation in the Drosophila brain induces oxidative stress by generating reactive oxygen species (ROS) and lipid peroxidation (LPO), depleting antioxidant enzymes. Molecular docking studies have evidenced it. Pb directly binds to antioxidants and major grooves of DNA, leading to DNA damage. Increased DNA damage, apoptosis, vacuolation in brains of Pb-treated ORR, Sod, or Cat knockdown flies; and on the contrary, reduced oxidative DNA damage, apoptosis, and vacuolation in brains of Pb treated Sod or Cat overexpressed flies put forward that oxidative stress is the mechanism in Pb caused neurotoxicity.


Assuntos
Encéfalo/efeitos dos fármacos , Chumbo/toxicidade , Síndromes Neurotóxicas/etiologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Encéfalo/fisiopatologia , Catalase/genética , Dano ao DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Drosophila melanogaster , Feminino , Técnicas de Silenciamento de Genes , Chumbo/administração & dosagem , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Simulação de Acoplamento Molecular , Síndromes Neurotóxicas/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo
13.
Oxid Med Cell Longev ; 2021: 6685204, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34336113

RESUMO

The developing brain is extremely sensitive to many chemicals. Perinatal exposure to neurotoxicants has been implicated in several neurodevelopmental disorders, including autism spectrum disorder, attention-deficit hyperactive disorder, and schizophrenia. Studies of the molecular and cellular events related to developmental neurotoxicity have identified a number of "adverse outcome pathways," many of which share oxidative stress as a key event. Oxidative stress occurs when the balance between the production of free oxygen radicals and the activity of the cellular antioxidant system is dysregulated. In this review, we describe some of the developmental neurotoxins that target the antioxidant system and the mechanisms by which they elicit stress, including oxidative phosphorylation in mitochondria and plasma membrane redox system in rodent models. We also discuss future directions for identifying adverse outcome pathways related to oxidative stress and developmental neurotoxicity, with the goal of improving our ability to quickly and accurately screen chemicals for their potential developmental neurotoxicity.


Assuntos
Síndromes Neurotóxicas/fisiopatologia , Estresse Oxidativo/imunologia , Animais , Feminino , Humanos , Masculino , Camundongos , Ratos
14.
Int J Mol Sci ; 22(15)2021 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-34360696

RESUMO

Neurodevelopment is uniquely sensitive to toxic insults and there are concerns that environmental chemicals are contributing to widespread subclinical developmental neurotoxicity (DNT). Increased DNT evaluation is needed due to the lack of such information for most chemicals in common use, but in vivo studies recommended in regulatory guidelines are not practical for the large-scale screening of potential DNT chemicals. It is widely acknowledged that developmental neurotoxicity is a consequence of disruptions to basic processes in neurodevelopment and that testing strategies using human cell-based in vitro systems that mimic these processes could aid in prioritizing chemicals with DNT potential. Myelination is a fundamental process in neurodevelopment that should be included in a DNT testing strategy, but there are very few in vitro models of myelination. Thus, there is a need to establish an in vitro myelination assay for DNT. Here, we summarize the routes of myelin toxicity and the known models to study this particular endpoint.


Assuntos
Encéfalo/fisiopatologia , Modelos Biológicos , Bainha de Mielina , Transtornos do Neurodesenvolvimento/etiologia , Síndromes Neurotóxicas/etiologia , Oligodendroglia , Animais , Humanos , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/fisiopatologia , Síndromes Neurotóxicas/fisiopatologia , Organoides , Testes de Toxicidade
15.
Life Sci ; 282: 119823, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34273375

RESUMO

AIMS: Zinc oxide nanoparticles (ZnO-NPs) are currently applied in food and pharmaceutical industries whose neurotoxic effect on the central nervous system (CNS) is a major concern. Considering the pharmacological properties (antioxidant, anti-inflammatory) of the geraniol (GE), we aimed to investigate the efficacy of geraniol on ZnO-NPs neurotoxicity. MATERIALS AND METHODS: We used 32 male Wistar rats, randomly assigned to four groups (n = 8): Control, GE (daily received 100 mg/kg of GE by gavage), ZnO-NPs (received intraperitoneal injection of 75 mg/kg of ZnO-NPs twice a week), and ZnO-NPs + GE (received both GE and ZnO-NPs at same doses above during 4 weeks). Morris water maze (MWM) and Y-maze tasks were done to evaluate learning and memory function. Biochemical assays were done to measure total antioxidant capacity (TAC), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPX) and ZnO-NPs bioaccumulation. Nissl and H&E staining were performed for histological evaluations. KEY FINDINGS: The results of behavioral study revealed that GE improved learning and memory impairment induced by ZnO-NPs. Moreover, neuroprotective effect of GE significantly decreased pathological parameters such as necrosis and gliosis, and consequently increased the number of nerve cells in the cortex and different hippocampal areas. Furthermore, biochemical studies demonstrated that GE significantly increased antioxidant indices (namely, TAC, SOD, and GPX) and reduced oxidative stress marker (MDA) and Zn bioaccumulation in ZnO-NPs treated animals. SIGNIFICANCE: Our results provide experimental evidence to further investigate the precise mechanisms underlying the geraniol as a promising therapeutic approach for improvement of cognitive function and neurotoxicity induce by ZnO-NPs.


Assuntos
Monoterpenos Acíclicos/farmacologia , Antioxidantes/farmacologia , Transtornos da Memória , Síndromes Neurotóxicas , Estresse Oxidativo/efeitos dos fármacos , Óxido de Zinco/toxicidade , Animais , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/fisiopatologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/fisiopatologia , Ratos , Ratos Wistar
16.
J Appl Toxicol ; 41(12): 2055-2067, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33993517

RESUMO

Silver nanoparticles (AgNPs) have become widespread in the environment with increasing industrial applications. But the studies about their potential health risks are far from enough, especially in neurotoxic effects. This study aimed to investigate the neurotoxic effects of longer-term exposure (prolonged exposure for 48 h and chronic exposure for 6 days) of 20nm AgNPs with/without polyvinylpyrrolidone (PVP) coating at low concentrations (0.01-10 mg·L-1 ) to Caenorhabditis elegans. The results suggested that exposure to AgNPs induced damage to nematode survival, with the longest and relative average life span reduced. Exposure to AgNPs caused neurotoxicity on locomotion behaviors (head thrashes, body bends, pharyngeal pumping frequency, and defecation interval) and sensory perception behaviors (chemotaxis assay and thermotaxis assay), as well as impaired dopaminergic, GABAergic, and cholinergic neurons, except for glutamatergic, based on the alters fluorescence intensity, in a dose- and time-dependent manner. Further investigations suggested that the low-dose AgNPs (0.01-0.1 mg·L-1 ) exposure raises receptors of GABAergic and dopamine in C. elegans at the genetic level, whereas opposite results were observed at higher doses (1-10 mg·L-1 ), which implied that AgNPs could cause neurotoxicity by impairing neurotransmitter delivery. The PVP-AgNPs could cause a higher fatality rate and neurotoxicity at the same dose. Notably, AgNPs did not cause any deleterious effect on nematodes at the lowest dose of 0.01 mg·L-1 . In general, these results suggested that AgNPs possess the neurotoxic potential in C. elegans and provided useful information to understand the neurotoxicity of AgNPs, which would offer an inspiring perspective on the safe application.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Neurônios/efeitos dos fármacos , Povidona/toxicidade , Prata/toxicidade , Animais , Caenorhabditis elegans/fisiologia , Neurônios/fisiologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Excipientes Farmacêuticos/toxicidade , Substitutos do Plasma/toxicidade
17.
Hum Exp Toxicol ; 40(11): 1962-1973, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34002645

RESUMO

The application of gold nanoparticle-peptide conjugates as theranostic agents for colorectal cancer shows much promise. This study aimed at determining the neurotoxic impact of 14 nm gold nanoparticles (AuNPs) functionalized with colorectal cancer-targeting peptides (namely p.C, p.L or p.14) in a rat model. Brain tissue samples, obtained from Wistar rats that received a single injection of citrate-capped AuNPs, polyethylene glycol-coated (PEG) AuNPs, p.C-PEG-AuNPs, p.L-PEG-AuNPs or p.14-PEG-AuNPs, and sacrificed after 2- and 12-weeks, respectively, were analysed. Inflammation marker (tumour necrosis factor-α, interleukin-6, interleukin-1ß), oxidative stress (superoxide dismutase, catalase, glutathione peroxidase) and apoptotic biomarker (cytochrome c, caspase-3) levels were measured. Gold nanoparticle-treated groups sacrificed after 2-weeks did not exhibit any significant inflammatory, oxidative stress or apoptotic effects in brain tissue compared to the untreated control group. In brain tissue from rats that were exposed to citrate-capped AuNPs for 12-weeks, tumour necrosis factor-α and interleukin-6 levels were significantly increased compared to the untreated control. Exposure to PEG-AuNP, p.C-PEG-AuNP, p.L-PEG-AuNP and p.14-PEG-AuNP did not elicit significant toxic effects compared to the control after 12-weeks, as evidenced by the absence of inflammatory, oxidative stress and apoptotic effects in brain tissue. We thus report on the safety of PEG-coated AuNP-peptide conjugates for potential application in the diagnosis of colorectal cancer; however, exposure to citrate-capped AuNPs could induce delayed neuro-inflammation, and as such, warrants further investigation.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Ouro/toxicidade , Ouro/uso terapêutico , Nanopartículas Metálicas/toxicidade , Nanopartículas Metálicas/uso terapêutico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Animais , Masculino , Terapia de Alvo Molecular/efeitos adversos , Ratos , Ratos Wistar
18.
Expert Opin Drug Saf ; 20(7): 827-838, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33789560

RESUMO

INTRODUCTION: We aimed to review cases of Syndrome of Irreversible Lithium-Effectuated Neurotoxicity (SILENT) characterized by neurological sequelae following acute lithium toxicity and to explore whether cerebellar sequelae are more frequent in cases presenting with fever and/or infection. AREAS COVERED: Case reports were identified from: (i) 6 reviews published up to 2005; (ii) MEDLINE, Web of Sciences, Cochrane Library and PsycINFO search. EXPERT OPINION: We identified 123 SILENT cases published from 1965 to 2019, in which cerebellar sequelae were observed in an overwhelming proportion (79%). SILENT may occur at any time during lithium treatment. This complication is most frequently observed during routine lithium treatment, with fewer than 10% of cases occurring after accidental or intentional overdoses. SILENT may occur even when lithium plasma levels are within the therapeutic range: 63% of cases had lithium plasma level <2.5 mEq/l (low/mild toxicity). Fever and/or infection were reported in nearly half of the patients (48%). The likelihood of presenting with cerebellar vs. other neurological sequelae was independently increased by elevated plasma lithium level (≥ 2.5 mEq/l) and by a history of fever and/or infection. Lithium users should be warned of the need to consult in case of fever to adjust their lithium dosage.


Assuntos
Antimaníacos/envenenamento , Compostos de Lítio/envenenamento , Síndromes Neurotóxicas/etiologia , Antimaníacos/administração & dosagem , Antimaníacos/farmacocinética , Relação Dose-Resposta a Droga , Overdose de Drogas , Febre/etiologia , Humanos , Compostos de Lítio/administração & dosagem , Compostos de Lítio/farmacocinética , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/fisiopatologia
19.
Int J Mol Sci ; 22(7)2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33805345

RESUMO

Acrylamide is a commonly used industrial chemical that is known to be neurotoxic to mammals. However, its developmental toxicity is rarely assessed in mammalian models because of the cost and complexity involved. We used zebrafish to assess the neurotoxicity, developmental and behavioral toxicity of acrylamide. At 6 h post fertilization, zebrafish embryos were exposed to four concentrations of acrylamide (10, 30, 100, or 300 mg/L) in a medium for 114 h. Acrylamide caused developmental toxicity characterized by yolk retention, scoliosis, swim bladder deficiency, and curvature of the body. Acrylamide also impaired locomotor activity, which was measured as swimming speed and distance traveled. In addition, treatment with 100 mg/L acrylamide shortened the width of the brain and spinal cord, indicating neuronal toxicity. In summary, acrylamide induces developmental toxicity and neurotoxicity in zebrafish. This can be used to study acrylamide neurotoxicity in a rapid and cost-efficient manner.


Assuntos
Acrilamida/toxicidade , Embrião não Mamífero/efeitos dos fármacos , Síndromes Neurotóxicas/fisiopatologia , Peixe-Zebra/crescimento & desenvolvimento , Acrilamida/farmacologia , Sacos Aéreos/patologia , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Embrião não Mamífero/fisiopatologia , Desenvolvimento Embrionário/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Escoliose/etiologia , Natação , Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...