Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 933
Filtrar
1.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38892402

RESUMEN

In day-to-day living, individuals are exposed to various environmentally hazardous substances that have been associated with diverse diseases. Exposure to air pollutants can occur during breathing, posing a considerable risk to those with environmental health vulnerabilities. Among vulnerable individuals, maternal exposure can negatively impact the mother and child in utero. The developing fetus is particularly vulnerable to environmentally hazardous substances, with potentially greater implications. Among air pollutants, toluene is neurotoxic, and its effects have been widely explored. However, the impact of low-level toluene exposure in daily life remains unclear. Herein, we evaluated 194 mothers and infants from the Growing children's health and Evaluation of Environment (GREEN) cohort to determine the possible effects of early-life toluene exposure on the nervous system. Using Omics experiments, the effects of toluene were confirmed based on epigenetic changes and altered mRNA expression. Various epigenetic changes were identified, with upregulated expression potentially contributing to diseases such as glioblastoma and Alzheimer's, and downregulated expression being associated with structural neuronal abnormalities. These findings were detected in both maternal and infant groups, suggesting that maternal exposure to environmental hazardous substances can negatively impact the fetus. Our findings will facilitate the establishment of environmental health policies, including the management of environmentally hazardous substances for vulnerable groups.


Asunto(s)
Exposición Materna , Tolueno , Humanos , Tolueno/toxicidad , Femenino , Lactante , Exposición Materna/efectos adversos , Embarazo , Adulto , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Sistema Nervioso/crecimiento & desarrollo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Epigénesis Genética/efectos de los fármacos , Masculino , Madres , Contaminantes Atmosféricos/toxicidad , Recién Nacido
2.
Sci Total Environ ; 869: 161738, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-36690096

RESUMEN

Evidence suggests that fluoride-induced neurodevelopment damage is linked to mitochondrial disorder, yet the detailed mechanism remains unclear. A cohort of Sprague-Dawley rats developmentally exposed to sodium fluoride (NaF) was established to simulate actual exposure of human beings. Using high-input proteomics and small RNA sequencing technology in rat hippocampus, we found mitochondrial translation as the most striking enriched biological process after NaF treatment, which involves the differentially expressed Required Meiotic Nuclear Division 1 homolog (RMND1) and neural-specific miR-221-3p. Further experiments in vivo and in vitro neuroendocrine pheochromocytoma (PC12) cells demonstrated that NaF impaired mitochondrial translation and function, as shown by declined mitochondrial membrane potential and inhibited expression of mitochondrial translation factors, mitochondrial translation products, and OXPHOS complexes, which was concomitant with decreased RMND1 and transcription factor c-Fos in mRNA and proteins as well as elevated miR-221-3p. Notably, RMND1 overexpression alleviated the NaF-elicited mitochondrial translation impairment by up-regulating translation factors, but not vice versa. Interestingly, ChIP-qPCR confirmed that c-Fos specifically controls the RMND1 transcription through direct binding with Rmnd1 promotor. Interference of gene expression verified c-Fos as an upstream positive regulator of RMND1, implicating in fluoride-caused mitochondrial translation impairment. Furthermore, dual-luciferase reporter assay evidenced that miR-221-3p targets c-Fos by binding its 3' untranslated region. By modulating the miR-221-3p expression, we identified miR-221-3p as a critical negative regulator of c-Fos. More importantly, we proved that miR-221-3p inhibitor improved mitochondrial translation and mitochondrial function to combat NaF neurotoxicity via activating the c-Fos/RMND1 axis, whereas miR-221-3p mimic tended towards opposite effects. Collectively, our data suggest fluoride impairs mitochondrial translation by dysregulating the miR-221-3p/c-Fos/RMND1 axis to trigger mitochondrial dysfunction, leading to neuronal death and neurodevelopment defects.


Asunto(s)
Fluoruros , MicroARNs , Trastornos del Neurodesarrollo , Animales , Humanos , Ratas , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/farmacología , Fluoruros/metabolismo , Fluoruros/toxicidad , MicroARNs/efectos de los fármacos , MicroARNs/genética , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/patología , Trastornos del Neurodesarrollo/inducido químicamente , Trastornos del Neurodesarrollo/metabolismo , Células PC12 , Ratas Sprague-Dawley , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo
3.
J Neuroinflammation ; 19(1): 289, 2022 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-36463233

RESUMEN

BACKGROUND: Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS: Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS: The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1ß, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS: Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Animales , Masculino , Ratones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/inmunología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Modelos Animales de Enfermedad , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/genética , Enfermedades Neuroinflamatorias/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/inmunología
4.
Reprod Biol Endocrinol ; 20(1): 19, 2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35081973

RESUMEN

BACKGROUND: Nitric oxide and GnRH are biological factors that participate in the regulation of reproductive functions. To our knowledge, there are no studies that link NO and GnRH in the sympathetic ganglia. Thus, the aim of the present work was to investigate the influence of NO on GnRH release from the coeliac ganglion and its effect on luteal regression at the end of pregnancy in the rat. METHODS: The ex vivo system composed by the coeliac ganglion, the superior ovarian nerve, and the ovary of rats on day 21 of pregnancy was incubated for 180 min with the addition, into the ganglionic compartment, of L-NG-nitro arginine methyl ester (L-NAME), a non-selective NO synthase inhibitor. The control group consisted in untreated organ systems. RESULTS: The addition of L-NAME in the coeliac ganglion compartment decreased NO as well as GnRH release from the coeliac ganglion. In the ovarian compartment, and with respect to the control group, we observed a reduced release of GnRH, NO, and noradrenaline, but an increased production of progesterone, estradiol, and expression of their limiting biosynthetic enzymes, 3ß-HSD and P450 aromatase, respectively. The inhibition of NO production by L-NAME in the coeliac ganglion compartment also reduced luteal apoptosis, lipid peroxidation, and nitrotyrosine, whereas it increased the total antioxidant capacity within the corpora lutea. CONCLUSION: Collectively, the results indicate that NO production by the coeliac ganglion modulates the physiology of the ovary and luteal regression during late pregnancy in rats.


Asunto(s)
Cuerpo Lúteo/inervación , Cuerpo Lúteo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Óxido Nítrico/metabolismo , Animales , Interacciones Farmacológicas , Femenino , Ganglios Simpáticos/efectos de los fármacos , Ganglios Simpáticos/metabolismo , Edad Gestacional , Hormona Liberadora de Gonadotropina/farmacología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/metabolismo , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Óxido Nítrico/farmacología , Ovario/inervación , Ovario/metabolismo , Embarazo , Ratas
5.
Acta Biochim Pol ; 68(4): 557-563, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34351731

RESUMEN

Benzophenone-3 (2-hydroxy-4-methoxybenzophenone, oxybenzone, or BP-3) is one of the most frequently used UV radiation absorbents, which are commonly referred to as sunscreen filters. Its widespread use in industrial applications provides protection against the photodegradation of a wide range of products but at the same time creates the risk of human exposure to benzophenone-3 unbeknownst to the individuals exposed. Topically applied benzophenone-3 penetrates individual skin layers, enters the bloodstream, and is excreted in the urine. In addition, benzophenone-3 easily crosses the placental barrier, which creates the risk of exposure to this substance in the prenatal period. Despite the widespread use and occurrence of benzophenone-3 in the human environment, little knowledge of the mechanisms underlying the effect of benzophenone-3 on the nervous system was available until recently. Only the most recent research, including studies by our group, has enabled the identification of new molecular mechanisms through which benzophenone-3 affects embryonic neuronal cells and the developing mammalian brain. Benzophenone-3 has been shown to induce neurotoxicity and apoptotic processes and inhibit autophagy in embryonic neuronal cells. Benzophenone-3 also alters expression and impairs function of receptors necessary for the proper development and function of the nervous system. The most worrying finding seems to be that benzophenone-3 contributes to an increased risk of developmental abnormalities and/or epigenetically based degeneration of neuronal cells by changing the epigenetic status of neuronal cells.


Asunto(s)
Benzofenonas/toxicidad , Sistema Nervioso/efectos de los fármacos , Protectores Solares/toxicidad , Rayos Ultravioleta , Administración Tópica , Benzofenonas/administración & dosificación , Sistema Endocrino/efectos de los fármacos , Exposición a Riesgos Ambientales , Humanos , Factores de Riesgo , Piel/efectos de los fármacos , Neoplasias Cutáneas/inducido químicamente , Protectores Solares/administración & dosificación
6.
Pharmacol Res ; 170: 105700, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34087351

RESUMEN

Cytisine, a natural bioactive compound that is mainly isolated from plants of the Leguminosae family (especially the seeds of Laburnum anagyroides), has been marketed in central and eastern Europe as an aid in the clinical management of smoking cessation for more than 50 years. Its main targets are neuronal nicotinic acetylcholine receptors (nAChRs), and pre-clinical studies have shown that its interactions with various nAChR subtypes located in different areas of the central and peripheral nervous systems are neuroprotective, have a wide range of biological effects on nicotine and alcohol addiction, regulate mood, food intake and motor activity, and influence the autonomic and cardiovascular systems. Its relatively rigid conformation makes it an attractive template for research of new derivatives. Recent studies of structurally modified cytisine have led to the development of new compounds and for some of them the biological activities are mediated by still unidentified targets other than nAChRs, whose mechanisms of action are still being investigated. The aim of this review is to describe and discuss: 1) the most recent pre-clinical results obtained with cytisine in the fields of neurological and non-neurological diseases; 2) the effects and possible mechanisms of action of the most recent cytisine derivatives; and 3) the main areas warranting further research.


Asunto(s)
Alcaloides/farmacología , Sistema Nervioso/efectos de los fármacos , Receptores Nicotínicos/efectos de los fármacos , Agentes para el Cese del Hábito de Fumar/farmacología , Cese del Hábito de Fumar , Alcaloides/farmacocinética , Alcaloides/toxicidad , Animales , Azocinas/farmacocinética , Azocinas/farmacología , Azocinas/toxicidad , Humanos , Estructura Molecular , Sistema Nervioso/metabolismo , Quinolizinas/farmacocinética , Quinolizinas/farmacología , Quinolizinas/toxicidad , Receptores Nicotínicos/metabolismo , Agentes para el Cese del Hábito de Fumar/farmacocinética , Agentes para el Cese del Hábito de Fumar/toxicidad , Relación Estructura-Actividad
7.
Ecotoxicol Environ Saf ; 217: 112198, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33862428

RESUMEN

The mechanism of neurodevelopmental toxicity of decabromodiphenyl ether (BDE209) remains unclear. Recent evidence suggests that neurosteroids disorders play a vital role in BDE209 induced-neurodevelopmental toxicity. To explore the mechanism of it, pregnant ICR mice were orally gavaged with 0, 225, and 900 mg kg-1 BDE209 for about 42 days. Spatial learning and memory abilities of offspring were tested on postnatal day (PND) 21. Offspring were euthanized at PND26, the neuronal structure, neurosteroids level, and related proteins including neurosteroids synthase, ionotropic receptors and cAMP-response element binding protein (CREB) pathway were evaluated, as well as Ca2+ concentration and the mitochondrial membrane potential (Mmp). Our results showed that BDE209 impaired learning and memory abilities and disrupted neuronal structure. Meanwhile, BDE209 decreased the pregnenolone (PREG), dehydroepiandrosterone (DHEA), progesterone (PROG) and allopregnanolone (ALLO) levels in the serum and brain, as well as the mRNA and protein levels of cholesterol-side-chain cleavage enzyme (P450scc), steroid 17α-hy-droxylase (P450C17), 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and steroid 5α-reductase of type I (5α-R) in the hippocampi. Also, BDE209 suppressed mRNA and protein levels of NR1, NR2A and NR2B subunits of the N-methyl-D-aspartic acid receptor (NMDAR) and α1 subunit of the Gamma-amino butyric acid A receptor (GABAAR), but increased the levels of ß2 and γ2 subunits of the GABAAR in the hippocampi. Moreover, BDE209 increased the Ca2+ concentration and phosphorylation extracellular regulated protein kinases (P-ERK) 1/2 level, but decreased the P-CREB and Mmp level in the hippocampi. These results indicate that BDE209 exposure during pregnancy and lactation is possible to affect learning and memory formation of offspring by the neurosteroid-mediated ionotropic receptors dysfunction.


Asunto(s)
Éteres Difenilos Halogenados/toxicidad , Sistema Nervioso/crecimiento & desarrollo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Lactancia , Masculino , Ratones , Ratones Endogámicos ICR , Sistema Nervioso/efectos de los fármacos , Neuronas/metabolismo , Neuroesteroides , Embarazo , Pregnanolona/metabolismo , Progesterona/metabolismo , Receptores de GABA/metabolismo , Esteroide 17-alfa-Hidroxilasa/metabolismo
8.
Food Chem Toxicol ; 151: 112134, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33762183

RESUMEN

T-2 toxin, a food-derived mycotoxin, has been identified as a neurotoxin. Nonetheless, T-2 toxin-induced neuroinflammation has never been revealed. As an important therapeutic target for inflammatory diseases and cancers, the role of high mobility group B1 (HMGB1) in mycotoxin-mediated neurotoxicity remains a mystery. In current study, we found that PC12 cells were sensitive to trace amounts of T-2 toxin less than 12 ng/mL, distinguished by decreased cell viability and increased release of lactate dehydrogenase (LDH). Oxidative stress and mitochondrial damage were observed in PC12 cells, manifested as accumulation of oxidative stress products, up-regulation of Nrf2/HO-1 pathway and decrease of mitochondrial membrane potential (MMP), leading to mitochondria-dependent apoptosis. Meanwhile, we first discovered that tiny amounts of T-2 toxin triggered neuroinflammation directly, including raising the expression and translocation of NF-κB and promoting secretion of proinflammatory cytokines such as TNF-α, IL-6, IL-8 and IL-1ß. Most interestingly, the increased of HMGB1 was detected both inside and outside the cells. Conversely, HMGB1 siRNA reduced T-2 toxin-mediated oxidative stress, apoptosis and neuroinflammatory outbreak, accompanied by lessened caspase-3 and caspase-9, and decreased secretion of pro-inflammatory cytokines. Taken together, T-2 toxin-stimulated PC12 cells simultaneously displayed apoptosis and inflammation, whereas HMGB1 played a critical role in these neurotoxic processes.


Asunto(s)
Proteína HMGB1/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Toxina T-2/toxicidad , Animales , Relación Dosis-Respuesta a Droga , FN-kappa B/metabolismo , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo
9.
Toxins (Basel) ; 13(2)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562446

RESUMEN

For thousands of years, Cannabis sativa has been utilized as a medicine and for recreational and spiritual purposes. Phytocannabinoids are a family of compounds that are found in the cannabis plant, which is known for its psychotogenic and euphoric effects; the main psychotropic constituent of cannabis is Δ9-tetrahydrocannabinol (Δ9-THC). The pharmacological effects of cannabinoids are a result of interactions between those compounds and cannabinoid receptors, CB1 and CB2, located in many parts of the human body. Cannabis is used as a therapeutic agent for treating pain and emesis. Some cannabinoids are clinically applied for treating chronic pain, particularly cancer and multiple sclerosis-associated pain, for appetite stimulation and anti-emesis in HIV/AIDS and cancer patients, and for spasticity treatment in multiple sclerosis and epilepsy patients. Medical cannabis varies from recreational cannabis in the chemical content of THC and cannabidiol (CBD), modes of administration, and safety. Despite the therapeutic effects of cannabis, exposure to high concentrations of THC, the main compound that is responsible for most of the intoxicating effects experienced by users, could lead to psychological events and adverse effects that affect almost all body systems, such as neurological (dizziness, drowsiness, seizures, coma, and others), ophthalmological (mydriasis and conjunctival hyperemia), cardiovascular (tachycardia and arterial hypertension), and gastrointestinal (nausea, vomiting, and thirst), mainly associated with recreational use. Cannabis toxicity in children is more concerning and can cause serious adverse effects such as acute neurological symptoms (stupor), lethargy, seizures, and even coma. More countries are legalizing the commercial production and sale of cannabis for medicinal use, and some for recreational use as well. Liberalization of cannabis laws has led to increased incidence of toxicity, hyperemesis syndrome, lung disease cardiovascular disease, reduced fertility, tolerance, and dependence with chronic prolonged use. This review focuses on the potential therapeutic effects of cannabis and cannabinoids, as well as the acute and chronic toxic effects of cannabis use on various body systems.


Asunto(s)
Cannabinoides/uso terapéutico , Cannabis , Marihuana Medicinal/uso terapéutico , Sistema Nervioso/efectos de los fármacos , Plantas Tóxicas , Animales , Cannabinoides/efectos adversos , Cannabinoides/aislamiento & purificación , Cannabis/efectos adversos , Humanos , Abuso de Marihuana/metabolismo , Abuso de Marihuana/fisiopatología , Abuso de Marihuana/psicología , Marihuana Medicinal/efectos adversos , Sistema Nervioso/metabolismo , Sistema Nervioso/fisiopatología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Síndromes de Neurotoxicidad/psicología , Plantas Tóxicas/efectos adversos , Receptores de Cannabinoides/efectos de los fármacos , Receptores de Cannabinoides/metabolismo , Transducción de Señal
11.
Anesth Analg ; 133(3): 595-605, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33497062

RESUMEN

BACKGROUND: Exposure to surgery and anesthesia in early childhood has been found to be associated with an increased risk of behavioral deficits. While the US Food and Drug Administration (FDA) has warned against prenatal exposure to anesthetic drugs, little clinical evidence exists to support this recommendation. This study evaluates the association between prenatal exposure to general anesthesia due to maternal procedures during pregnancy and neuropsychological and behavioral outcome scores at age 10. METHODS: This is an observational cohort study of children born in Perth, Western Australia, with 2 generations of participants contributing data to the Raine Study. In the Raine Study, the first generation (Gen1) are mothers enrolled during pregnancy, and the second generation (Gen2) are the children born to these mothers from 1989 to 1992 with neuropsychological and behavioral tests at age 10 (n=2024). In the primary analysis, 6 neuropsychological and behavioral tests were evaluated at age 10: Raven's Colored Progressive Matrices (CPM), McCarron Assessment of Neuromuscular Development (MAND), Peabody Picture Vocabulary Test (PPVT), Symbol Digit Modality Test (SDMT) with written and oral scores, Clinical Evaluation of Language Fundamentals (CELF) with Expressive, Receptive, and Total language scores, and Child Behavior Checklist (CBCL) with Internalizing, Externalizing, and Total behavior scores. Outcome scores of children prenatally exposed to general anesthesia were compared to children without prenatal exposure using multivariable linear regression models adjusting for demographic and clinical covariates (sex, race, income, and maternal education, alcohol or tobacco use, and clinical diagnoses: diabetes, epilepsy, hypertension, psychiatric disorders, or thyroid dysfunction). Bonferroni adjustment was used for the 6 independent tests in the primary analysis, so a corrected P value <.0083 (P = .05 divided by 6 tests, or a 99.17% confidence interval [CI]) was required for statistical significance. RESULTS: Among 2024 children with available outcome scores, 22 (1.1%) were prenatally exposed to general anesthesia. Prenatally exposed children had higher CBCL Externalizing behavioral scores (score difference of 6.1 [99.17% CI, 0.2-12.0]; P = .006) than unexposed children. Of 6 tests including 11 scores and subscores, only CBCL Externalizing behavioral scores remained significant after multiple comparisons adjustment with no significant differences found in any other score. CONCLUSIONS: Prenatal exposure to general anesthetics is associated with increased externalizing behavioral problems in childhood. However, given the limitations of this study and that avoiding necessary surgery during pregnancy can have significant detrimental effects on the mother and the child, further studies are needed before changes to clinical practice are made.


Asunto(s)
Anestesia General/efectos adversos , Anestésicos Generales/efectos adversos , Trastornos de la Conducta Infantil/inducido químicamente , Conducta Infantil/efectos de los fármacos , Desarrollo Infantil/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Niño , Trastornos de la Conducta Infantil/diagnóstico , Trastornos de la Conducta Infantil/fisiopatología , Trastornos de la Conducta Infantil/psicología , Femenino , Humanos , Masculino , Sistema Nervioso/crecimiento & desarrollo , Embarazo , Medición de Riesgo , Factores de Riesgo , Australia Occidental
12.
Eur J Pharmacol ; 895: 173870, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33476653

RESUMEN

Fibromyalgia is a potentially disabling chronic disease, characterized by widespread pain and a range of comorbidities such as hypertension. Among the mechanisms involved in fibromyalgia-like pain symptoms are kinins and their B1 and B2 receptors. Moreover, angiotensin I converting enzyme (ACE) inhibitors, commonly used as antihypertensive drugs, can enhance pain by blocking the degradation of peptides such as substance P and bradykinin, besides enhancing kinin receptors signalling. We investigated the effect of ACE inhibitors on reserpine-induced fibromyalgia-like pain symptoms and the involvement of kinins in this effect in mice. Nociceptive parameters (mechanical and cold allodynia and overt nociception) were evaluated after ACE inhibitors administration in mice previously treated with reserpine. The role of kinin B1 and B2 receptors was investigated using pharmacological antagonism. Additionally, bradykinin levels, as well as the activity of ACE and kininase I, were measured in the sciatic nerve, spinal cord and cerebral cortex of the mice. The ACE inhibitors enalapril and captopril enhanced reserpine-induced mechanical allodynia, and this increase was prevented by kinin B1 and B2 receptor antagonists. Substance P and bradykinin caused overt nociception and increased mechanical allodynia in animals treated with reserpine. Reserpine plus ACE inhibitors increased bradykinin-related peptide levels and inhibited ACE activity in pain modulation structures. Since hypertension is a frequent comorbidity affecting fibromyalgia patients, hypertension treatment with ACE inhibitors in these patients should be reviewed once this could enhance fibromyalgia-like pain symptoms. Thus, the treatment of hypertensive patients with fibromyalgia could include other classes of antihypertensive drugs, different from ACE inhibitors.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/toxicidad , Fibromialgia/inducido químicamente , Sistema Nervioso/efectos de los fármacos , Dolor Nociceptivo/inducido químicamente , Umbral del Dolor/efectos de los fármacos , Peptidil-Dipeptidasa A/metabolismo , Receptores de Bradiquinina/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Captopril/toxicidad , Modelos Animales de Enfermedad , Enalapril/toxicidad , Fibromialgia/enzimología , Fibromialgia/fisiopatología , Masculino , Ratones , Sistema Nervioso/enzimología , Sistema Nervioso/fisiopatología , Dolor Nociceptivo/enzimología , Dolor Nociceptivo/fisiopatología , Reserpina , Transducción de Señal
13.
Ann Rev Mar Sci ; 13: 137-160, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32749906

RESUMEN

Millions of tons of oil are spilled in aquatic environments every decade, and this oil has the potential to greatly impact fish populations. Here, we review available information on the physiological effects of oil and polycyclic aromatic hydrocarbons on fish. Oil toxicity affects multiple biological systems, including cardiac function, cholesterol biosynthesis, peripheral and central nervous system function, the stress response, and osmoregulatory and acid-base balance processes. We propose that cholesterol depletion may be a significant contributor to impacts on cardiac, neuronal, and synaptic function as well as reduced cortisol production and release. Furthermore, it is possible that intracellular calcium homeostasis-a part of cardiotoxic and neuronal function that is affected by oil exposure-may be related to cholesterol depletion. A detailed understanding of oil impacts and affected physiological processes is emerging, but knowledge of their combined effects on fish in natural habitats is largely lacking. We identify key areas deserving attention in future research.


Asunto(s)
Peces/fisiología , Contaminación por Petróleo/análisis , Petróleo/toxicidad , Hidrocarburos Policíclicos Aromáticos/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Ecosistema , Corazón/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Petróleo/análisis , Hidrocarburos Policíclicos Aromáticos/análisis , Contaminantes Químicos del Agua/análisis
14.
Phytother Res ; 35(6): 2997-3012, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33368772

RESUMEN

Sweet almond (Prunus dulcis (Mill.) D.A.Webb) is a known nut, which has long been used in several ethnomedical systems, especially in Persian medicine (PM) for its nutritional and therapeutic activities. In this study, we aimed to provide a summary on traditional uses, phytochemistry, and pharmacological activities of sweet almond. Thus, we reviewed textbooks of PM and electronic literature on traditional medicine. Moreover, the available data on the usage of sweet almond were searched in electronic databases to find articles on its pharmacological properties and phytochemistry. According to phytochemical investigations, this plant contains macronutrients, micronutrients, essential oils, various phenolic compounds, and phytosterols. Current pharmacological studies represent that Prunus dulcis has several biological activities including prebiotic, antimicrobial, antioxidant, antiinflammatory, anticancer, hepatoprotective, cardiometabolic protection, nootropic, anxiolytic, sedative-hypnotic, and nervous-improving effects. Further clinical trials and meta-analysis are required to draw a definitive conclusion on the efficacy and therapeutic activities of almond.


Asunto(s)
Medicina Tradicional , Nueces/química , Fitoterapia , Extractos Vegetales/farmacología , Prunus dulcis/química , Animales , Antiinfecciosos/farmacología , Antiinfecciosos/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Suplementos Dietéticos/análisis , Humanos , Sistema Nervioso/efectos de los fármacos , Aceites Volátiles/farmacología , Persia , Fenoles/farmacología , Fenoles/uso terapéutico , Fitosteroles/farmacología , Fitosteroles/uso terapéutico
15.
Neurotoxicology ; 83: 166-178, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33290785

RESUMEN

The European Union's REACH Regulation requires determination of potential health and environmental effects of chemicals in commerce. The present case study examines the application of REACH guidance for health hazard assessments of three high production volume (HPV) aluminium (Al) substances: metallic aluminium, aluminium oxide, and aluminium hydroxide. Among the potential adverse health consequences of aluminium exposure, neurotoxicity is one of the most sensitive targets of Al toxicity and the most critical endpoint. This case study illustrates integration of data from multiple lines of evidence into REACH weight of evidence evaluations. This case study then explains how those results support regulatory decisions on classification and labelling. Challenges in the REACH appraisal of Al compounds include speciation, solubility and bioavailability, application of assessment factors, read-across rationale and differences with existing regulatory standards. Lessons learned from the present case study relate to identification and evaluation of toxicologic and epidemiologic data; assessing data relevance and reliability; development of derived no-effect levels (DNELs); addressing data gaps and preparation of chemical safety reports.


Asunto(s)
Hidróxido de Aluminio/toxicidad , Óxido de Aluminio/toxicidad , Aluminio/toxicidad , Sistema Nervioso/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Pruebas de Toxicidad , Aluminio/farmacocinética , Hidróxido de Aluminio/farmacocinética , Óxido de Aluminio/farmacocinética , Animales , Europa (Continente) , Unión Europea , Humanos , Sistema Nervioso/metabolismo , Sistema Nervioso/patología , Sistema Nervioso/fisiopatología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/fisiopatología , Medición de Riesgo , Toxicocinética
16.
Ecotoxicol Environ Saf ; 209: 111832, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33383341

RESUMEN

Cobalt has been known for its neurotoxicity in numerous studies. However, the molecular mechanism underlying cobalt-induced neurotoxicity remains largely unknown. In this study, two neuroblastoma (SHSY5Y and N2a) cell lines and a phaeochromocytoma (PC12) line were used as in vitro models. Cells were treated for 24 h with 50, 100, 200, 300, 400 µM cobalt chloride (CoCl2) or cultured with 300 µM CoCl2 for 4, 8, 12 and 24 h to investigate the effects of histone acetylation on CoCl2-induced neurodegenerative damages. Our findings demonstrate that CoCl2 suppresses the acetylation of histone H3 and H4 in a time-dependent and dosage-dependent manner. Furthermore, CoCl2 selectively decreases the expression and activity of histone acetyltransferase (HAT) but has no effects on histone deacetylase (HDAC) in SHSY5Y cells. More importantly, we show that 100 ng/mL HDAC inhibitor trichostatin (TSA) pre-treatment partly attenuates 300 µM CoCl2-induced neurodegenerative damages in SHSY5Y cells. Mechanistic analyses show that CoCl2-induced neurodegenerative damages are associated with the dysfunction of APP, BACE1, PSEN1, NEP and HIF-1α genes, whose expression are partly mediated by histone modification. In summary, we demonstrate that histone acetylation is involved in CoCl2-induced neurodegenerative damages. Our study indicates an important connection between histone modification and the pathological process of neurodegenerative damages and provides a mechanism for cobalt-mediated epigenetic regulation.


Asunto(s)
Cobalto/toxicidad , Histonas/fisiología , Sistema Nervioso/efectos de los fármacos , Acetilación/efectos de los fármacos , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular Tumoral , Cobalto/metabolismo , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Pruebas de Toxicidad
17.
J Appl Toxicol ; 41(8): 1241-1261, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33184925

RESUMEN

QXOH-Levobupivacaine (LB) is a fixed-dose combination of 35-mM QXOH and 10-mM LB. It was developed for perioperative analgesia because of its long-acting analgesic effect. The purpose of this study was to evaluate the potential toxicity of QXOH-LB in beagle dogs in accordance with the Guidance on the repeated-dose toxicity published by the China Food and Drug Administration. Groups of five male and five female beagle dogs received normal saline, QXOH-LB (2, 4, and 8 mg/kg, calculated as QXOH), QXOH (2, 4, and 8 mg/kg), or LB (2 mg/kg, equals the concentration of LB in 8-mg/kg QXOH-LB group) at the volume of 1 mL/kg once per day for 14 days through subcutaneous injection. No mortality was observed. Dogs in the control group as well as animals treated with 2-mg/kg QXOH or QXOH-LB exhibited normal behaviors. Clinical signs of toxicity in dogs treated with 4 and 8 mg/kg of QXOH or QXOH-LB included decreased activity, unsteady gait, jerks, tremors, vocalization, emesis, ataxia, lateral/sternal recumbency, deep/rapid respiration, and gasping. Additionally, neurological function was found to be affected by QXOH and QXOH-LB at the doses of 4 and 8 mg/kg. All clinical signs were recovered within 24 h. The no-observed-adverse-effect level of QXOH and QXOH-LB was considered to be 2 mg/kg. Toxicokinetic data showed that exposure to QXOH and LB increased as QXOH-LB doses were increased from 4 to 8 mg/kg. There was no evidence of drug accumulation or any effect of gender.


Asunto(s)
Anestésicos Locales/toxicidad , Levobupivacaína/toxicidad , Lidocaína/análogos & derivados , Anestésicos Locales/administración & dosificación , Animales , Coagulación Sanguínea/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Perros , Combinación de Medicamentos , Electrocardiografía/efectos de los fármacos , Femenino , Levobupivacaína/administración & dosificación , Lidocaína/administración & dosificación , Lidocaína/toxicidad , Masculino , Sistema Nervioso/efectos de los fármacos , Frecuencia Respiratoria/efectos de los fármacos
18.
Rev Chil Anest ; 50(4): 576-581, 2021.
Artículo en Español | LILACS | ID: biblio-1526286

RESUMEN

Total intravenous anesthesia (TIVA) with propofol/remifentanil appears in the literatura as a good option for neurosurgical patients who have increased intracranial pressure (ICP),risk of postoperative nausea and vomiting (PONV), need for neuromonitoring, and in those with impaired brain self-regulation. On the other hand, in patients with normal neurological status, normal ICP, a technique with volatile (halogenated) agents plus an opiiid can be used. This review describes two anesthetic techniques available for use in neurosurgery, highlighting the neurophysiological changes, advantages and disadvantages of each technique. MATERIAL AND METHOD: PubMed search engine was used for bibliographic search. DISCUSSION: The search for an ideal anesthetic in neurosurgery is still a matter of debate. There are numerous investigations aimed at finding an optimal agent that ensure the coupling between cerebral flow (CBF) and metabolism, keeping self-regulation intact without increasing the CBF and intracerebral pressure (ICP). CONCLUSIONS: Both anesthetic techniques, TIVA and volatile agents (halogenated), can be used in neurosurgical procedures and should provide neuroprotection, brain relaxation and a rapid awakening.


La anestesia total endovenosa (TIVA) con propofol/remifentanilo aparece en la literatura como una buena opción para pacientes neuroquirúrgicos que tienen aumento de la presión intracraneana (PIC), riesgo de náuseas y vómitos posoperatorios (NVPO), necesidad de neuromonitoreo, y en aquellos con alteración de la autorregulación cerebral. Por otra parte, en pacientes con estado neurológico normal, PIC normal puede usarse una técnica con agentes volátiles (halogenados) más un opioide. Esta revisión describe dos técnicas anestésicas disponibles para su uso en neurocirugía, destaca los cambios neurofisiológicos, ventajas y desventajas de cada técnica. MATERIAL Y MÉTODO: Para búsqueda bibliográfica se usó buscador PubMed. DISCUSIÓN: La búsqueda de un anestésico ideal en neurocirugía sigue siendo tema de debate. Existen numerosas investigaciones destinadas a buscar un agente óptimo que asegure el acoplamiento entre flujo sanguíneo cerebral (FSC) y metabolismo, manteniendo la autorregulación intacta sin aumentar el FSC y presión intracerebral (PIC). CONCLUSIONES: Ambas técnicas anestésicas, TIVA y agentes volátiles (halogenados), pueden ser usadas en procedimientos neuroquirúrgicos y deben brindar neuroprotección, relajación cerebral y un despertar rápido.


Asunto(s)
Humanos , Procedimientos Neuroquirúrgicos/métodos , Anestesia por Inhalación/métodos , Anestesia Intravenosa/métodos , Anestésicos Intravenosos/efectos adversos , Anestésicos Intravenosos/farmacología , Anestésicos por Inhalación/efectos adversos , Anestésicos por Inhalación/farmacología , Náusea y Vómito Posoperatorios/inducido químicamente , Neuroprotección , Sistema Nervioso/efectos de los fármacos
19.
Molecules ; 25(22)2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33187049

RESUMEN

Flavonoids are phytochemical compounds present in many plants, fruits, vegetables, and leaves, with potential applications in medicinal chemistry. Flavonoids possess a number of medicinal benefits, including anticancer, antioxidant, anti-inflammatory, and antiviral properties. They also have neuroprotective and cardio-protective effects. These biological activities depend upon the type of flavonoid, its (possible) mode of action, and its bioavailability. These cost-effective medicinal components have significant biological activities, and their effectiveness has been proved for a variety of diseases. The most recent work is focused on their isolation, synthesis of their analogs, and their effects on human health using a variety of techniques and animal models. Thousands of flavonoids have been successfully isolated, and this number increases steadily. We have therefore made an effort to summarize the isolated flavonoids with useful activities in order to gain a better understanding of their effects on human health.


Asunto(s)
Flavonoides/química , Flavonoides/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antifúngicos/química , Antifúngicos/farmacología , Antimaláricos/química , Antimaláricos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Antivirales/química , Antivirales/farmacología , Sistema Cardiovascular/efectos de los fármacos , Flavonoides/economía , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Ratones , Sistema Nervioso/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Hojas de la Planta/química , Plantas/química , Polifenoles/química , Polifenoles/farmacología , Quercetina/química , Quercetina/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/prevención & control
20.
Molecules ; 25(21)2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-33167585

RESUMEN

Neurodegenerative diseases (ND) can be characterized by degradation and subsequent loss of neurons. ND has been identified as the leading cause of disability-adjusted life years (DALYs) worldwide and is associated with various risk factors such as ageing, certain genetic polymorphisms, inflammation, immune and metabolic conditions that may induce elevated reactive oxygen species (ROS) release and subsequent oxidative stress. Presently, no specific cure or prevention is available for ND patients; the symptoms can be only alleviated via drug treatment or surgery. The existing pharmacological treatments are only available for partial treatment of the symptoms. A natural product known as oil palm phenolics (OPP), which is high in antioxidant, could become a potential supplementary antioxidant for neurodegenerative health. OPP is a water-soluble extract from palm fruit that demonstrated medicinal properties including anti-tumor, anti-diabetic and neuroprotective effects. In this review, OPP was proposed for its neuroprotective effects via several mechanisms including antioxidant and anti-inflammatory properties. Besides, OPP has been found to modulate the genes involved in neurotrophic activity. The evidence and proposed mechanism of OPP on the neuroprotective health may provide a comprehensive natural medicine approach to alleviate the symptoms of neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas/tratamiento farmacológico , Aceite de Palma/farmacología , Fenoles/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Radicales Libres , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Inflamación , Sistema Nervioso/efectos de los fármacos , Neuronas/metabolismo , Neuroprotección , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo , Enfermedad de Parkinson/tratamiento farmacológico , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA