Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Chemosphere ; 349: 140991, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38141683

RESUMEN

BACKGROUND: Coronary atherosclerotic disease (CAD) is often accompanied by peripheral atherosclerosis, resulting in a higher risk of ischemia and cardiovascular death. Exposure to metals is associated with atherosclerotic plaques at specific sites. However, less is known about the effects of mixed metals on systemic atherosclerotic burden in CAD patients. OBJECTIVES: To investigate the association of metal mixtures with systemic atherosclerotic burden in a CAD population. METHODS: A cross-sectional study including 1562 CAD patients from Southwest China was conducted. The levels of 10 blood metals were measured via inductively coupled plasma spectrometry. More than one vessel with a stenosis ≥50% vessel diameter was defined as CAD. Carotid and lower limb atherosclerosis was assessed by using ultrasound, and coronary atherosclerosis was quantified via arterial angiography. Systemic atherosclerosis was scored according to the presence or absence of lesions at the three sites and the total number of lesions. To investigate the combined impacts and interaction effects of metals, Bayesian kernel machine regression was used. Weighted quantile regression was used to identify the contributions of the metals. RESULTS: Significant overall associations of mixed metals with systemic atherosclerotic burden were found. These positive overall associations were mainly driven by Cd, Cu and Pb in systemic atherosclerosis. The main contributing factors were As and Cu for coronary atherosclerosis as well as Cd, Cu and Pb for carotid and lower limb atherosclerosis. Cd and Pb or Cr can interact, and Pb interacts with age, sex and alcohol. CONCLUSIONS: In CAD patients, exposure to combinations of metals was highly positively associated with systemic atherosclerotic burden. These significant trends were more pronounced in the peripheral arteries and carotid arteries. Controlling environmental metal exposure can contribute to reducing systemic atherosclerosis in CAD patients.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Humanos , Enfermedad de la Arteria Coronaria/epidemiología , Enfermedad de la Arteria Coronaria/patología , Estudios Transversales , Teorema de Bayes , Cadmio , Plomo , Aterosclerosis/epidemiología , Factores de Riesgo
2.
Ecotoxicol Environ Saf ; 263: 115241, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37441943

RESUMEN

Cadmium (Cd) exposure is a risk factor for endothelial dysfunction and cardiovascular disease. Ferroptosis is a type of cell death that relies on lipid peroxidation. Whether ferroptosis acts in Cd-induced vascular endothelial damage and the underlying mechanisms remain unclear. Herein, we found that Cd resulted in ferroptosis of vascular endothelial cells (ECs) in vivo and in vitro. In the visualized zebrafish embryos, Cd accumulated in vascular ECs, ROS and lipid peroxidation levels were increased, and the oxidoreductase system was disturbed after exposure. Moreover, Cd decreased Gpx4 in ECs and caused smaller mitochondria with increased membrane density. Accompanied by ferroptosis, the number of ECs and the area of the caudal venous plexus in zebrafish embryos were reduced, and the survival rate of HUVECs decreased. These effects were partially reversed by ferrostatin-1 and aggravated by erastin. Mechanistically, an excessive increase in Heat Shock Protein 70 (Hsp70) was identified by transcriptomics after Cd exposure. Inhibition of Hsp70 by VER-155008 or siRNA ameliorated Cd-induced ferroptosis, thereby alleviating endothelial injury. Furthermore, Hsp70 regulated Cd-induced ferroptosis by targeting multiple targets, including Gpx4, Fth1, Nrf2 and Acsl4. Our findings provide a new approach to investigating the endothelial damage of Cd and indicate that regulation of Hsp70 is an important target for alleviating this process.


Asunto(s)
Ferroptosis , Proteínas HSP70 de Choque Térmico , Animales , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Pez Cebra/metabolismo , Cadmio/metabolismo , Células Endoteliales/metabolismo
4.
J Hazard Mater ; 453: 131369, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37086674

RESUMEN

Paraquat (PQ) is the most widely used herbicide in agriculture worldwide and has been considered a high-risk environmental factor for Parkinson's disease (PD). Chronic PQ exposure selectively induces dopaminergic neuron loss, the hallmark pathologic feature of PD, resulting in Parkinson-like movement disorders. However, the underlying mechanisms remain unclear. Here, we demonstrated that repetitive PQ exposure caused dopaminergic neuron loss, dopamine deficiency and motor deficits dose-dependently in mice. Accordingly, mitochondrial calcium uniporter (MCU) was highly expressed in PQ-exposed mice and neuronal cells. Importantly, MCU knockout (KO) effectively rescued PQ-induced dopaminergic neuron loss and motor deficits in mice. Genetic and pharmacological inhibition of MCU alleviated PQ-induced mitochondrial dysfunction and neuronal death in vitro. Mechanistically, PQ exposure triggered mitochondrial fragmentation via imbalance of the optic atrophy 1 (OPA1) processing manifested by cleavage of L-OPA1 to S-OPA1, which was reversed by inhibition of MCU. Notably, the upregulation of MCU was mediated by miR-129-1-3p posttranscriptionally, and overexpression of miR-129-1-3p could rebalance OPA1 processing and attenuate mitochondrial dysfunction and neuronal death induced by PQ exposure. Consequently, our work uncovers an essential role of MCU and a novel molecular mechanism, miR-MCU-OPA1, in PQ-induced pathogenesis of PD, providing a potential target and strategy for environmental neurotoxins-induced PD treatment.


Asunto(s)
MicroARNs , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Parkinson/genética , Regulación hacia Arriba , Paraquat/toxicidad , Dopamina
5.
Adv Sci (Weinh) ; 10(7): e2203869, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36642847

RESUMEN

Cadmium (Cd) is a high-risk pathogenic toxin for hepatic diseases. Excessive mitophagy is a hallmark in Cd-induced hepatotoxicity. However, the underlying mechanism remains obscure. Mitochondrial calcium uniporter (MCU) is a key regulator for mitochondrial and cellular homeostasis. Here, Cd exposure upregulated MCU expression and increased mitochondrial Ca2+ uptake are found. MCU inhibition through siRNA or by Ru360 significantly attenuates Cd-induced excessive mitophagy, thereby rescues mitochondrial dysfunction and increases hepatocyte viability. Heterozygous MCU knockout mice exhibit improved liver function, ameliorated pathological damage, less mitochondrial fragmentation, and mitophagy after Cd exposure. Mechanistically, Cd upregulates MCU expression through phosphorylation activation of cAMP-response element binding protein at Ser133(CREBS133 ) and subsequent binding of MCU promoter at the TGAGGTCT, ACGTCA, and CTCCGTGATGTA regions, leading to increased MCU gene transcription. The upregulated MCU intensively interacts with voltage-dependent anion-selective channel protein 1 (VDAC1), enhances its dimerization and ubiquitination, resulting in excessive mitophagy. This study reveals a novel mechanism, through which Cd upregulates MCU to enhance mitophagy and hepatotoxicity.


Asunto(s)
Cadmio , Canales de Calcio , Enfermedad Hepática Inducida por Sustancias y Drogas , Proteínas Mitocondriales , Mitofagia , Canal Aniónico 1 Dependiente del Voltaje , Animales , Ratones , Cadmio/toxicidad , Canales de Calcio/genética , Canales de Calcio/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Dimerización , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mitofagia/genética , Mitofagia/fisiología , Ubiquitinación , Regulación hacia Arriba , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
6.
Environ Int ; 169: 107512, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36108500

RESUMEN

Paraquat (PQ) is the most widely used herbicide in the world and a well-known potent neurotoxin for humans. PQ exposure has been linked to increase the risk of Parkinson's disease (PD). However, the mechanism underlying its neurotoxic effects in PD pathogenesis is unclear. In our present study, C57BL/6J mice treated with PQ manifested severe motor deficits indicated by the significant reductions in suspension score, latency to fall from rotarod, and grip strength at 8 weeks after PQ exposure. Pathological hallmarks of Parkinsonism in the midbrain such as dopaminergic neuron loss, increased α-synuclein protein, and dysregulated PD-related genes were observed. Non-targeted lipidome analysis demonstrated that PQ exposure alters lipid profile and abundance, increases pro-inflammatory lipids.27 significantly altered subclasses of lipids belonged to 6 different lipid categories. Glycerophospholipids, sphingolipids, and glycerides were the most abundant lipids. Abundance of pro-inflammatory lipids such as Cer, LPC, LPS, and LPI was significantly increased in the midbrain. mRNA expressions of genes regulating ceramide biosynthesis in the midbrain were markedly up-regulated. Moreover, PQ exposure increased serum pro-inflammatory cytokines and provoked neuroinflammation in the midbrain. Pro-inflammatory lipids and cytokines in the midbrain were positively correlated with motor deficits. PQ poisoning in humans significantly also elevated serum pro-inflammatory cytokines and induced an intense systemic inflammation. In summary, we presented initial investigations of PQ induced molecular events related to the PD pathogenesis, capturing aspects of disturbed lipid metabolism, neuroinflammation, impairment of dopaminergic neurons in the midbrain, and an intense systemic inflammation. These neurotoxic effects of PQ exposure may mechanistically contribute to the pathogenesis of PQ induced Parkinsonism. Results of this study also strongly support the hypothesis that ever-increasing prevalence of Parkinson's disease is etiologically linked to the health risk of exposure to neurotoxic environmental pollutants.


Asunto(s)
Contaminantes Ambientales , Herbicidas , Síndromes de Neurotoxicidad , Enfermedad de Parkinson , Trastornos Parkinsonianos , Animales , Ceramidas/farmacología , Citocinas , Contaminantes Ambientales/toxicidad , Glicéridos/farmacología , Glicerofosfolípidos/farmacología , Herbicidas/toxicidad , Humanos , Lipopolisacáridos/farmacología , Mesencéfalo , Ratones , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias , Síndromes de Neurotoxicidad/etiología , Neurotoxinas , Paraquat/toxicidad , Enfermedad de Parkinson/etiología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/complicaciones , ARN Mensajero , Esfingolípidos/farmacología , alfa-Sinucleína/farmacología
7.
Toxicology ; 453: 152726, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33617915

RESUMEN

Cadmium (Cd) is a ubiquitous environmental and occupational pollutant that is considered as a high-risk factor for neurodegenerative diseases. However, the mechanism underlying Cd-induced neurotoxicity has not been fully elucidated. Abnormal mitochondrial distribution and excessive mitochondrial fission are increasingly implicated in various neurological pathologies. Herein, by exposing primary cortical neurons to Cd (10 and 100 µM) for various times (0, 6, 12, and 24 h), we observed that the rapid motility of the mitochondria in neurons progressively slowed. Many more mitochondria were transported and distributed to the somas of Cd-treated neurons. Coupled with abnormal mitochondrial distribution, Cd exposure triggered excessive mitochondrial fragmentation, followed by mitochondrial membrane potential loss and neuronal damage. However, BAPTA-AM, a chelator of cytosolic calcium ([Ca2+]c), significantly attenuated Cd-induced abnormal mitochondrial distribution and excessive mitochondrial fission, which protected against Cd-induced mitochondrial damage and neuronal toxicity. In contrast to the increase in [Ca2+]c, Cd exposure had no effect on the level of mitochondrial calcium ([Ca2+]m). Inhibiting [Ca2+]m uptake, either by ruthenium 360 (Ru360) or by knock-out of mitochondrial calcium uniporter (MCU), failed to alleviate Cd-induced mitochondrial damage and neuronal toxicity. Additionally, in MCU knock-out neurons, BAPTA-AM effectively prevented Cd-induced abnormal mitochondrial distribution and excessive mitochondrial fission. Taken together, Cd exposure disrupts mitochondrial distribution and activates excessive mitochondrial fission by elevating [Ca2+]c independent of MCU-mediated mitochondrial calcium uptake, thereby leading to neurotoxicity. Chelating overloaded [Ca2+]c is a promising strategy to prevent the neurotoxicity of Cd.


Asunto(s)
Cadmio/toxicidad , Canales de Calcio/deficiencia , Calcio/metabolismo , Citosol/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales/fisiología , Animales , Animales Recién Nacidos , Canales de Calcio/genética , Células Cultivadas , Citosol/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
8.
Environ Pollut ; 258: 113319, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31882189

RESUMEN

Thallium (TI) is one of the most toxic heavy metals and priority pollutant metals. The emerging TI environmental pollution worldwide has posed a great threat to human health. However, based on the World Health Organization (WHO), the risk and severity of adverse health effects of TI in the range of 5-500 µg/L are uncertain. Moreover, evidence regarding the adverse impacts of TI on children's health is still insufficient. Herein, we aim to investigate the early adverse effects of TI on children's health and provide references for the WHO to establish stricter safety limits of TI. From 2015 to 2019, urinary TI and many clinical laboratory parameters related to blood routine, hepatic, renal, myocardial, coagulation function and serum electrolyte were measured in six children aged 1-9 years. The urinary TI concentration ranged from 13.4 µg/L to 60.1 µg/L with a mean of 36.1 µg/L and a median of 34.8 µg/L in six children in 2015. Although only four children felt a little poor appetite, several laboratory abnormalities indicated early damage in liver, renal, and myocardial functions in all children in 2015. After treatment and following up for four years, although the children's TI concentration decreased below 5 µg/L, their liver and renal functions did not completely recover, and their myocardial function worsened. Results indicated that impaired liver, renal, and myocardial functions were closely associated with elevated urinary TI concentration in children. Considering the increasing use of TI in high-technology industries and emerging TI environmental-contamination zones worldwide, establishing stricter safety limits of TI and paying more attention to the adverse health effects of TI on children are urgently required. SUMMARY: We found that a relatively low concentration of thallium (13.4 µg/L to 60.1 µg/L) impaired liver, renal, and myocardial function in six children. After treatment and following up these children for four years, although their urinary TI concentration decreased below 5 µg/L, their liver and renal functions did not completely recover, and their myocardial function worsened.


Asunto(s)
Contaminantes Ambientales/orina , Corazón/fisiopatología , Riñón/fisiopatología , Hígado/fisiopatología , Metales Pesados/orina , Talio/efectos adversos , Talio/orina , Contaminantes Químicos del Agua/orina , Niño , Preescolar , Exposición a Riesgos Ambientales , Femenino , Humanos , Lactante , Masculino , Metales Pesados/toxicidad , Infarto del Miocardio , Contaminantes Químicos del Agua/toxicidad
9.
BMJ Open ; 9(4): e026638, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975682

RESUMEN

OBJECTIVE: To investigate the extent and associations of patient/diagnostic delay and other potential factors with catastrophic health expenditure (CHE) for tuberculosis (TB) care in Chongqing municipality, China. DESIGN: A cross-sectional study. SETTING: Four counties of Chongqing municipality, China. PARTICIPANTS: A total of 1199 patients with active pulmonary TB beyond 16 years and without mental disorders were consecutively recruited in the four counties' designated TB medical institutions. OUTCOME MEASURES: The incidence and intensity of CHE for TB care were described. The association between patients' 'sociodemographic and clinical characteristics such as patient delay, diagnostic delay, forms of TB, health insurance status and hospitalisation and CHE were analysed using univariate and multivariate logistic regression. RESULTS: The incidence of CHE was 52.8% and out-of-pocket (OOP) payments were 93% of the total costs for TB care. Compared with patients without delay, the incidence and intensity of CHE were higher in patients who had patient delay or diagnostic delay. Patients who experienced patient delay or diagnostic delay, who was a male, elderly (≥60 years), an inhabitant, a peasant, divorced/widow, the New Cooperative Medical Scheme membership had greater risks of incurring CHE for TB care. Having a higher educational level appeared to be a protective factor. However, hospitalisation was not associated with CHE after controlling for other variables. CONCLUSION: The incidence and intensity of CHE for TB care are high, which provides baseline data about catastrophic costs that TB-related households faced in Chongqing of China. Variety of determinants of CHE implicate that it is essential to take effective measures to promote early seeking care and early diagnosis, improve the actual reimbursement rates of health insurance, especially for outpatients, and need more fine-tuned interventions such as precise poverty alleviation to reduce catastrophic costs of the vulnerable population.


Asunto(s)
Enfermedad Catastrófica/economía , Gastos en Salud , Tuberculosis/economía , Adolescente , Adulto , Enfermedad Catastrófica/epidemiología , China/epidemiología , Ciudades , Estudios Transversales , Diagnóstico Tardío , Femenino , Humanos , Incidencia , Seguro de Salud , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Salud Urbana , Adulto Joven
10.
PLoS One ; 11(3): e0150923, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26950212

RESUMEN

Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Campos Electromagnéticos , Células Madre Embrionarias/citología , Células-Madre Neurales/citología , Neuritas/efectos de la radiación , Canales Catiónicos TRPC/genética , Regulación hacia Arriba/efectos de la radiación , Animales , Encéfalo/embriología , Encéfalo/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Madre Embrionarias/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Células-Madre Neurales/efectos de la radiación , Neuritas/metabolismo , ARN Interferente Pequeño/genética , Canales Catiónicos TRPC/deficiencia
11.
Autophagy ; 11(7): 1037-51, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26120888

RESUMEN

Cadmium is one of the most toxic metal compounds found in the environment. It is well established that Cd induces hepatotoxicity in humans and multiple animal models. Melatonin, a major secretory product of the pineal gland, has been reported to protect against Cd-induced hepatotoxicity. However, the mechanism behind this protection remains to be elucidated. We exposed HepG2 cells to different concentrations of cadmium chloride (2.5, 5, and 10 µM) for 12 h. We found that Cd induced mitochondrial-derived superoxide anion-dependent autophagic cell death. Specifically, Cd decreased SIRT3 protein expression and activity and promoted the acetylation of SOD2, superoxide dismutase 2, mitochondrial, thus decreasing its activity, a key enzyme involved in mitochondrial ROS production, although Cd did not disrupt the interaction between SIRT3 and SOD2. These effects were ameliorated by overexpression of SIRT3. However, a catalytic mutant of SIRT3 (SIRT3(H248Y)) lacking deacetylase activity lost the capacity to suppress Cd-induced autophagy. Notably, melatonin treatment enhanced the activity but not the expression of SIRT3, decreased the acetylation of SOD2, inhibited mitochondrial-derived O2(•-) production and suppressed the autophagy induced by 10 µM Cd. Moreover, 3-(1H-1,2,3-triazol-4-yl)pyridine, a confirmed selective SIRT3 inhibitor, blocked the melatonin-mediated suppression of autophagy by inhibiting SIRT3-SOD2 signaling. Importantly, melatonin suppressed Cd-induced autophagic cell death by enhancing SIRT3 activity in vivo. These results suggest that melatonin exerts a hepatoprotective effect on mitochondrial-derived O2(•-)-stimulated autophagic cell death that is dependent on the SIRT3/SOD2 pathway.


Asunto(s)
Autofagia/efectos de los fármacos , Cadmio/toxicidad , Melatonina/farmacología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/metabolismo , Superóxido Dismutasa/metabolismo , Acetilación/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Citoprotección/efectos de los fármacos , Células Hep G2 , Humanos , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Modelos Biológicos , Piridinas/farmacología , Superóxidos/metabolismo
12.
Toxicol Appl Pharmacol ; 286(2): 80-91, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25840356

RESUMEN

With application of nano-sized nickel-containing particles (Nano-Ni) expanding, the health concerns about their adverse effects on the pulmonary system are increasing. However, the mechanisms for the pulmonary toxicity of these materials remain unclear. In the present study, we focused on the impacts of NiO nanoparticles (NiONPs) on sirtuin1 (SIRT1), a NAD-dependent deacetylase, and investigated whether SIRT1 was involved in NiONPs-induced apoptosis. Although the NiONPs tended to agglomerate in fluid medium, they still entered into the human bronchial epithelial cells (BEAS-2B) and released Ni(2+) inside the cells. NiONPs at doses of 5, 10, and 20µg/cm(2) inhibited the cell viability. NiONPs' produced cytotoxicity was demonstrated through an apoptotic process, indicated by increased numbers of Annexin V positive cells and caspase-3 activation. The expression of SIRT1 was markedly down-regulated by the NiONPs, accompanied by the hyperacetylation of p53 (tumor protein 53) and overexpression of Bax (Bcl-2-associated X protein). However, overexpression of SIRT1 through resveratrol treatment or transfection clearly attenuated the NiONPs-induced apoptosis and activation of p53 and Bax. Our results suggest that the repression of SIRT1 may underlie the NiONPs-induced apoptosis via p53 hyperacetylation and subsequent Bax activation. Because SIRT1 participates in multiple biologic processes by deacetylation of dozens of substrates, this knowledge of the impact of NiONPs on SIRT1 may lead to an improved understanding of the toxic mechanisms of Nano-Ni and provide a molecular target to antagonize Nano-Ni toxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Bronquios/metabolismo , Células Epiteliales/metabolismo , Nanopartículas/toxicidad , Níquel/toxicidad , Sirtuina 1/antagonistas & inhibidores , Bronquios/citología , Bronquios/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Humanos , Nanopartículas/metabolismo , Níquel/metabolismo , Sirtuina 1/genética
13.
Radiat Res ; 183(3): 305-14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25688995

RESUMEN

Extremely low-frequency electromagnetic fields (ELF-EMF) and radiofrequency electromagnetic fields (RF-EMF) have been considered to be possibly carcinogenic to humans. However, their genotoxic effects remain controversial. To make experiments controllable and results comparable, we standardized exposure conditions and explored the potential genotoxicity of 50 Hz ELF-EMF and 1800 MHz RF-EMF. A mouse spermatocyte-derived GC-2 cell line was intermittently (5 min on and 10 min off) exposed to 50 Hz ELF-EMF at an intensity of 1, 2 or 3 mT or to RF-EMF in GSM-Talk mode at the specific absorption rates (SAR) of 1, 2 or 4 W/kg. After exposure for 24 h, we found that neither ELF-EMF nor RF-EMF affected cell viability using Cell Counting Kit-8. Through the use of an alkaline comet assay and immunofluorescence against γ-H2AX foci, we found that ELF-EMF exposure resulted in a significant increase of DNA strand breaks at 3 mT, whereas RF-EMF exposure had insufficient energy to induce such effects. Using a formamidopyrimidine DNA glycosylase (FPG)-modified alkaline comet assay, we observed that RF-EMF exposure significantly induced oxidative DNA base damage at a SAR value of 4 W/kg, whereas ELF-EMF exposure did not. Our results suggest that both ELF-EMF and RF-EMF under the same experimental conditions may produce genotoxicity at relative high intensities, but they create different patterns of DNA damage. Therefore, the potential mechanisms underlying the genotoxicity of different frequency electromagnetic fields may be different.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Daño del ADN/efectos de la radiación , Campos Electromagnéticos , Ondas de Radio , Animales , Humanos , Masculino , Ratones , Espermatocitos/metabolismo , Espermatocitos/efectos de la radiación
14.
Am J Transl Res ; 7(11): 2364-78, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26807184

RESUMEN

Numerous analyses including in vivo and in vitro experiments have demonstrated that inhalation exposure of NiONPs can result in pulmonary fibrosis. However, the potential mechanisms of this pathological process remain elusive. Here, we investigate the role of HIF-1α and TGF-ß1 in NiONPs-induced pulmonary fibrosis with a focus on the interplay of the above two proteins. In vivo, male Sprague&Dawley rats were exposed to NiONPs and pulmonary fibrosis was demonstrated using H&E staining and immunochemistry of αSMA. In vitro, NiONPs contributed to cell proliferation and increased expressions of collagen-1 and αSMA in human fetal lung fibroblasts. Both HIF-1α and TGF-ß1 were upregulated by NiONPs treatment. Inhibition of HIF-1α reduced TGF-ß1 expression and downregulation of TGF-ß1 reduced HIF-1α protein level. Mechanism investigation revealed that TGF-ß1 affects nuclear translocation activity of HIF-1α. Taken together, these finding provide evidence that HIF-1α and TGF-ß1 act in synergy to foster NiONPs-induced pulmonary fibrosis, and the cross talk between them is a pivotal mechanism of pulmonary fibrosis.

15.
Toxicol Sci ; 142(1): 182-95, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25159133

RESUMEN

Melatonin is an indolamine synthesized in the pineal gland that has a wide range of physiological functions, and it has been under clinical investigation for expanded applications. Increasing evidence demonstrates that melatonin can ameliorate cadmium-induced hepatotoxicity. However, the potentially protective effects of melatonin against cadmium-induced hepatotoxicity and the underlying mechanisms of this protection remain unclear. This study investigates the protective effects of melatonin pretreatment on cadmium-induced hepatotoxicity and elucidates the potential mechanism of melatonin-mediated protection. We exposed HepG2 cells to different concentrations of cadmium chloride (2.5, 5, and 10 µM) for 12 h. We found that Cd stimulated cytotoxicity, disrupted the mitochondrial membrane potential, increased reactive oxygen species production, and decreased mitochondrial mass and mitochondrial DNA content. Consistent with this finding, Cd exposure was associated with decreased Sirtuin 1 (SIRT1) protein expression and activity, thus promoted acetylation of PGC-1 alpha, a key enzyme involved in mitochondrial biogenesis and function, although Cd did not disrupt the interaction between SIRT1 and PGC-1 alpha. However, all cadmium-induced mitochondrial oxidative injuries were efficiently attenuated by melatonin pretreatment. Moreover, Sirtinol and SIRT1 siRNA each blocked the melatonin-mediated elevation in mitochondrial function by inhibiting SIRT1/ PGC-1 alpha signaling. Luzindole, a melatonin receptor antagonist, was found to partially block the ability of melatonin to promote SIRT1/ PGC-1 alpha signaling. In summary, our results indicate that SIRT1 plays an essential role in the ability of moderate melatonin to stimulate PGC-1 alpha and improve mitochondrial biogenesis and function at least partially through melatonin receptors in cadmium-induced hepatotoxicity.


Asunto(s)
Cloruro de Cadmio/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Melatonina/farmacología , Mitocondrias/efectos de los fármacos , Receptor de Melatonina MT1/biosíntesis , Sirtuina 1/biosíntesis , Factores de Transcripción/biosíntesis , Supervivencia Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma
16.
Sci Rep ; 4: 5103, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24869783

RESUMEN

A radiofrequency electromagnetic field (RF-EMF) of 1800 MHz is widely used in mobile communications. However, the effects of RF-EMFs on cell biology are unclear. Embryonic neural stem cells (eNSCs) play a critical role in brain development. Thus, detecting the effects of RF-EMF on eNSCs is important for exploring the effects of RF-EMF on brain development. Here, we exposed eNSCs to 1800 MHz RF-EMF at specific absorption rate (SAR) values of 1, 2, and 4 W/kg for 1, 2, and 3 days. We found that 1800 MHz RF-EMF exposure did not influence eNSC apoptosis, proliferation, cell cycle or the mRNA expressions of related genes. RF-EMF exposure also did not alter the ratio of eNSC differentiated neurons and astrocytes. However, neurite outgrowth of eNSC differentiated neurons was inhibited after 4 W/kg RF-EMF exposure for 3 days. Additionally, the mRNA and protein expression of the proneural genes Ngn1 and NeuroD, which are crucial for neurite outgrowth, were decreased after RF-EMF exposure. The expression of their inhibitor Hes1 was upregulated by RF-EMF exposure. These results together suggested that 1800 MHz RF-EMF exposure impairs neurite outgrowth of eNSCs. More attention should be given to the potential adverse effects of RF-EMF exposure on brain development.


Asunto(s)
Campos Electromagnéticos , Células Madre Embrionarias/efectos de la radiación , Células-Madre Neurales/efectos de la radiación , Neurogénesis/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Teléfono Celular , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Ratones , Neuritas/efectos de la radiación , ARN Mensajero/biosíntesis , ARN Mensajero/efectos de la radiación , Ondas de Radio
17.
Cell Tissue Res ; 355(1): 223-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24085620

RESUMEN

Whether environmental exposure to bisphenol A (BPA) may induce reproductive disorders is still controversial but certain studies have reported that BPA may cause meiotic abnormalities in C. elegans and female mice. However, little is known about the effect of BPA on meiosis in adult males. To determine whether BPA exposure at an environmentally relevant dose could induce meiotic abnormalities in adult male rats, we exposed 9-week-old male Wistar rats to BPA by gavage at 20 µg/kg body weight (bw)/day for 60 consecutive days. We found that BPA significantly increased the proportion of stage VII seminiferous epithelium and decreased the proportion of stage VIII. Consequently, spermiation was inhibited and spermatogenesis was disrupted. Further investigation revealed that BPA exposure delayed meiosis initiation in the early meiotic stage and induced the accumulation of chromosomal abnormalities and meiotic DNA double-strand breaks (DSBs) in the late meiotic stage. The latter event subsequently activated the phosphatidylinositol 3-kinase-related protein kinase (ATM). Our results suggest that long-term exposure to BPA may lead to continuous meiotic abnormalities and ultimately put mammalian reproductive health at risk.


Asunto(s)
Contaminantes Ocupacionales del Aire/efectos adversos , Compuestos de Bencidrilo/efectos adversos , Estrógenos no Esteroides/efectos adversos , Meiosis/efectos de los fármacos , Fenoles/efectos adversos , Epitelio Seminífero/efectos de los fármacos , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Compuestos de Bencidrilo/administración & dosificación , Roturas del ADN de Doble Cadena/efectos de los fármacos , Estrógenos no Esteroides/administración & dosificación , Masculino , Fenoles/administración & dosificación , Ratas , Ratas Wistar , Epitelio Seminífero/ultraestructura , Transducción de Señal/efectos de los fármacos , Espermatogénesis/efectos de los fármacos
18.
Bioelectromagnetics ; 35(1): 58-69, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24122970

RESUMEN

The constant exposure of modern society to extremely low frequency magnetic fields (ELF-MF) has raised considerable concerns about the potential risks to male reproduction. However, the epidemiological and experimental data remain contradictory and inconclusive. In the present study, we investigated the effects of 50 Hz ELF-MF of 500 µT applied 4 h/day, 7 days/week for 4 and 8 weeks on male reproduction, focusing on changes in spermatogenesis. Several biological endpoints related to testicular function and spermatogenesis were measured, including the following: body mass, masses of testes and epididymis, sperm count and abnormal sperm ratio in the caudal epididymis, serum testosterone level, testicular histology, frequency of 14 stages of the cycle of the seminiferous epithelium and of four stages of meiosis I, germ cell apoptosis and testicular oxidative status. No significant differences were found in the biological endpoints between the sham control and the exposed rats in either the 4- or 8-week exposure period. These negative results may result from the lack of change in serum testosterone. In conclusion, our study indicates that exposure to low intensity ELF-MF may have no adverse effects on spermatogenesis.


Asunto(s)
Campos Magnéticos , Espermatogénesis/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Peso Corporal/efectos de la radiación , Epidídimo/patología , Epidídimo/fisiología , Epidídimo/efectos de la radiación , Etiquetado Corte-Fin in Situ , Campos Magnéticos/efectos adversos , Masculino , Meiosis/efectos de la radiación , Tamaño de los Órganos , Estrés Oxidativo/efectos de la radiación , Distribución Aleatoria , Ratas Sprague-Dawley , Espermatogénesis/fisiología , Espermatozoides/efectos de la radiación , Testículo/patología , Testículo/fisiología , Testículo/efectos de la radiación , Testosterona/sangre
19.
Mutat Res ; 752(1-2): 57-67, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23402883

RESUMEN

Bisphenol A (BPA) is a well-known endocrine-disrupting chemical (EDC) that has received particular attention because of its widespread distribution in humans. Due to its chemical similarity to diethylstilbestrol, which is carcinogenic to mammals, the possible genotoxicity of BPA has already largely been evaluated. However, the results are still inconclusive and controversial. To investigate the genotoxic effects of BPA in rat germ cells and the potential protective action of melatonin against these effects, adult male Sprague-Dawley rats were orally administered BPA at a dose of 200mg/kg body weight per day for ten consecutive days with or without melatonin pretreatment. The thiobarbituric acid reactive substances (TBARS) level and superoxide dismutase (SOD) activity in the testes were evaluated. Subsequently, their spermatocytes were isolated, and DNA damage was assessed using an alkaline comet assay and the meiotic spread method. BPA administration did not significantly affect the weights of rats and their reproductive organs, and no alteration in sperm count was found. However, we demonstrated that BPA administration induced a significant increase in TBARS levels and a decrease in SOD activity that were concomitant with an increase in DNA migration within male germ cells and γH2AX foci formation on the autosomes of pachytene spermatocytes. Furthermore, a decrease in the proportion of 4C-cells was observed. These BPA effects were significantly alleviated by melatonin pretreatment. Nevertheless, the genotoxic effects of BPA were not accompanied by apoptosis in germ cells and morphological changes in the testes. These results indicate that BPA exposure may induce DNA damage accumulation in germ cells via oxidative stress. Moreover, melatonin may be a promising pharmacological candidate for preventing the potential genotoxicity of BPA following occupational or environmental exposure.


Asunto(s)
Antioxidantes/farmacología , Compuestos de Bencidrilo/toxicidad , Daño del ADN/efectos de los fármacos , Melatonina/farmacología , Mutágenos/toxicidad , Fenoles/toxicidad , Espermatozoides/efectos de los fármacos , Animales , Compuestos de Bencidrilo/antagonistas & inhibidores , Masculino , Estrés Oxidativo , Fenoles/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
20.
Toxicol Lett ; 218(1): 2-9, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23333639

RESUMEN

Whether exposure to radiofrequency electromagnetic radiation (RF-EMR) emitted from mobile phones can induce DNA damage in male germ cells remains unclear. In this study, we conducted a 24h intermittent exposure (5 min on and 10 min off) of a mouse spermatocyte-derived GC-2 cell line to 1800 MHz Global System for Mobile Communication (GSM) signals in GSM-Talk mode at specific absorption rates (SAR) of 1 W/kg, 2 W/kg or 4 W/kg. Subsequently, through the use of formamidopyrimidine DNA glycosylase (FPG) in a modified comet assay, we determined that the extent of DNA migration was significantly increased at a SAR of 4 W/kg. Flow cytometry analysis demonstrated that levels of the DNA adduct 8-oxoguanine (8-oxoG) were also increased at a SAR of 4 W/kg. These increases were concomitant with similar increases in the generation of reactive oxygen species (ROS); these phenomena were mitigated by co-treatment with the antioxidant α-tocopherol. However, no detectable DNA strand breakage was observed by the alkaline comet assay. Taking together, these findings may imply the novel possibility that RF-EMR with insufficient energy for the direct induction of DNA strand breaks may produce genotoxicity through oxidative DNA base damage in male germ cells.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , ADN/efectos de la radiación , Microondas/efectos adversos , Estrés Oxidativo/efectos de la radiación , Espermatocitos/efectos de la radiación , Animales , Antioxidantes/farmacología , Biomarcadores/metabolismo , Línea Celular , Supervivencia Celular/efectos de la radiación , Ensayo Cometa , Relación Dosis-Respuesta en la Radiación , Guanina/análogos & derivados , Guanina/metabolismo , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Oxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Espermatocitos/metabolismo , Espermatocitos/patología , alfa-Tocoferol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...