Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Ecotoxicol Environ Saf ; 279: 116481, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38788562

RESUMEN

Manganese (Mn) overexposure has been associated with the development of neurological damage reminiscent of Parkinson's disease, while the underlying mechanisms have yet to be fully characterized. This study aimed to investigate the mechanisms leading to injury in dopaminergic neurons induced by Mn and identify novel treatment approaches. In the in vivo and in vitro models, ICR mice and dopaminergic neuron-like PC12 cells were exposed to Mn, respectively. We treated them with anti-ferroptotic agents ferrostatin-1 (Fer-1), deferoxamine (DFO), HIF-1α activator dimethyloxalylglycine (DMOG) and inhibitor LW6. We also used p53-siRNA to verify the mechanism underlying Mn-induced neurotoxicity. Fe and Mn concentrations increased in ICR mice brains overexposed to Mn. Additionally, Mn-exposed mice exhibited movement impairment and encephalic pathological changes, with decreased HIF-1α, SLC7A11, and GPX4 proteins and increased p53 protein levels. Fer-1 exhibited protective effects against Mn-induced both behavioral and biochemical changes. Consistently, in vitro, Mn exposure caused ferroptosis-related changes and decreased HIF-1α levels, all ameliorated by Fer-1. Upregulation of HIF-1α by DMOG alleviated the Mn-associated ferroptosis, while LW6 exacerbated Mn-induced neurotoxicity through downregulating HIF-1α. p53 knock-down also rescued Mn-induced ferroptosis without altering HIF-1α protein expression. Mn overexposure resulted in ferroptosis in dopaminergic neurons, mediated through the HIF-1α/p53/SLC7A11 pathway.


Asunto(s)
Sistema de Transporte de Aminoácidos y+ , Encéfalo , Ferroptosis , Subunidad alfa del Factor 1 Inducible por Hipoxia , Manganeso , Ratones Endogámicos ICR , Proteína p53 Supresora de Tumor , Animales , Ferroptosis/efectos de los fármacos , Células PC12 , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Manganeso/toxicidad , Encéfalo/efectos de los fármacos , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Ratas , Masculino , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Ciclohexilaminas/farmacología , Fenilendiaminas/toxicidad , Fenilendiaminas/farmacología , Deferoxamina/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Aminoácidos Dicarboxílicos
2.
New Phytol ; 240(1): 173-190, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37563927

RESUMEN

The anther tapetum helps control microspore release and essential components for pollen wall formation. TAPETAL DEVELOPMENT and FUNCTION1 (TDF1) is an essential R2R3 MYB tapetum transcription factor in Arabidopsis thaliana; however, little is known about pollen development in the temperate monocot barley. Here, we characterize the barley (Hordeum vulgare L.) TDF1 ortholog using reverse genetics and transcriptomics. Spatial/temporal expression analysis indicates HvTDF1 has tapetum-specific expression during anther stage 7/8. Homozygous barley hvtdf1 mutants exhibit male sterility with retarded tapetum development, delayed tapetum endomitosis and cell wall degeneration, resulting in enlarged, vacuolated tapetum surrounding collapsing microspores. Transient protein expression and dual-luciferase assays show TDF1 is a nuclear-localized, transcription activator, that directly activates osmotin proteins. Comparison of hvtdf1 transcriptome data revealed several pathways were delayed, endorsing the observed retarded anther morphology. Arabidopsis tdf1 mutant fertility was recovered by HvTDF1, supporting a conserved role for TDF1 in monocots and dicots. This indicates that tapetum development shares similarity between monocot and dicots; however, barley HvTDF1 appears to uniquely act as a modifier to activate tapetum gene expression pathways, which are subsequently also induced by other factors. Therefore, the absence of HvTDF1 results in delayed developmental progression rather than pathway failure, although inevitably still results in pollen degeneration.


Asunto(s)
Arabidopsis , Hordeum , Hordeum/genética , Hordeum/metabolismo , Regulación de la Expresión Génica de las Plantas , Flores/fisiología , Arabidopsis/metabolismo , Factores de Transcripción/metabolismo
3.
J Arthroplasty ; 38(12): 2638-2643, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37286058

RESUMEN

BACKGROUND: The aim of this study was to compare outcomes after total hip arthroplasty (THA) in patients who have preoperative asymptomatic gluteal tendinosis (aGT) to a control group with no gluteal tendinosis (GT). METHODS: A retrospective analysis was performed using data from patients who underwent THA between March 2016 and October 2020. An aGT was diagnosed using hip magnetic resonance imaging (MRI) without clinical symptoms. Patients who had aGT were matched (1:1) to patients without GT on MRI. A total of 56 aGT hips and 56 hips without GT were found using propensity-score matching. Patient-reported outcomes, intraoperative macroscopic evaluation, outcome measurements, postoperative physical examinations, complications, and revisions were compared for both groups. RESULTS: When compared to preoperative outcomes, both groups demonstrated significant improvements in patients-reported outcomes at the final follow-up. There were no significant differences between both groups for preoperative scores, 2-year postoperative outcome scores, or the magnitude of improvement. Patients in the aGT group were significantly less likely to obtain the MCID for the SF-36 MCS score (50.2 versus 69.3%, P = .034). However, there were no other differences in the rates of meeting the MCID between both groups. The aGT group demonstrated significantly higher rates of partial tendon degeneration of the gluteus medius muscle. CONCLUSION: Asymptomatic gluteal tendinosis patients who have osteoarthritis and undergo THA may expect favorable patients-reported outcomes at minimum 2-year follow-up. These results were comparable with those of a control group of patients without gluteal tendinosis. LEVEL OF EVIDENCE: III.


Asunto(s)
Artroplastia de Reemplazo de Cadera , Tendinopatía , Humanos , Artroplastia de Reemplazo de Cadera/métodos , Estudios Retrospectivos , Músculo Esquelético/cirugía , Tendones/cirugía , Tendinopatía/cirugía , Resultado del Tratamiento , Articulación de la Cadera/diagnóstico por imagen , Articulación de la Cadera/cirugía , Medición de Resultados Informados por el Paciente
4.
J Foot Ankle Surg ; 62(2): 398-404, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36588066

RESUMEN

Regarding the treatment of ankle arthritis, the choice of arthroscopic ankle arthrodesis (AAA) or open ankle arthrodesis (OAA) remains controversial. To guide clinical decision-making, we conducted a meta-analysis on the optimal treatment of ankle arthrodesis. We identified eligible studies published from June 1, 1969 to June 1, 2020 using the Cochrane Library, PubMed, OVID, Embase, and Medline searched the references of relevant studies. Randomized and non-randomized studies that compared outcomes of AAA and OAA were included. After the methodologic assessment, available data were extracted and statistically reviewed. The primary outcomes were overall complications rate, tourniquet time, length of the hospital stay, non-union rate, and rate to fusion. The secondary outcomes were delayed union and postoperative infection rate. We included 9 studies comparing arthroscopic and open in patients with ankle arthrodesis, comprising 467 participants. AAA had the advantage of demonstrating a lower overall complication rate (odds ratio [OR], 0.44 [95% confidence interval [CI], 0.26-0.73]; p = .002), shorter intraoperative tourniquet time (mean difference [MD], -16.49 [95% CI, -23.51 to -9.46]; p < .001), shorter length of the hospital stay (MD -1.75, 95% CI -1.94 to -1.2, p < .001),lower non-union rate (OR, -0.07 [95% CI, -0.13 to -0.02]; p <.01) and higher rate to fusion (OR, 4.2 [95% CI, 1.96-8.99]; p < .001) in comparison with OAA. Yet, no significant differences were found in delayed union (OR, 0.46 [95% CI, 0.10-2.04]; p = .30) and postoperative infection rate (OR, 0.45 [95% CI, 0.17-1.15]; p = .09) between the groups. Our results suggest that arthroscopic ankle arthrodesis is superior to open ankle arthrodesis alone in the treatment of ankle arthritis based on the overall complication rate, intraoperative tourniquet time, length of the hospital stay, non-union rate and rate to fusion. However, further high-quality randomized controlled trials with appropriate blinding methods are needed to confirm the findings.


Asunto(s)
Tobillo , Artritis , Humanos , Tobillo/cirugía , Articulación del Tobillo/cirugía , Resultado del Tratamiento , Artroscopía/métodos , Estudios Retrospectivos , Artritis/cirugía , Artrodesis/métodos , Complicaciones Posoperatorias
5.
Drug Chem Toxicol ; 44(2): 130-139, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31258002

RESUMEN

Diabetes mellitus is a significant global public health issue. The diabetic state not only precipitates chronic disease but also has the potential to change the toxicity of drugs and chemicals. Acrylonitrile (AN) is a potent neurotoxin widely used in industrial products. This study used a streptozotocin (STZ)-induced diabetic rat model to examine the role of cytochrome P450 2E1 (CYP2E1) in acute AN toxicity. The protective effect of phenethyl isothiocyanate (PEITC), a phytochemical inhibitor of CYP2E1, was also investigated. A higher incidence of convulsions and loss of the righting reflex, and decreased rates of survival, as well as elevated CYP2E1 activity, were observed in diabetic rats treated with AN when compared to those in non-diabetic rats, suggesting that diabetes confers susceptibility to the acute toxicity of AN. Pretreatment with PEITC (20-80 mg/kg) followed by AN injection alleviated the acute toxicity of AN in diabetic rats as evidenced by the decreased incidence of convulsions and loss of righting reflex, and increased rates of survival. PEITC pretreatment at 40 and 80 mg/kg decreased hepatic CYP2E1 activity in AN-exposed diabetic rats. PEITC pretreatment (20 mg/kg) increased the glutathione (GSH) content and glutathione S-transferase (GST) activity and further decreased ROS levels in AN-exposed diabetic rats. Collectively, STZ-induced diabetic rats were more sensitive to AN-induced acute toxicity mainly due to CYP2E1 induction, and PEITC pretreatment significantly alleviated the acute toxicity of AN in STZ-induced diabetic rats. PEITC might be considered as a potential effective chemo-preventive agent against AN-induced acute toxicity in individuals with an underlying diabetic condition.


Asunto(s)
Acrilonitrilo/toxicidad , Inhibidores del Citocromo P-450 CYP2E1/farmacología , Diabetes Mellitus Experimental/fisiopatología , Isotiocianatos/farmacología , Animales , Citocromo P-450 CYP2E1/efectos de los fármacos , Citocromo P-450 CYP2E1/metabolismo , Inhibidores del Citocromo P-450 CYP2E1/administración & dosificación , Relación Dosis-Respuesta a Droga , Isotiocianatos/administración & dosificación , Masculino , Ratas , Ratas Sprague-Dawley , Reflejo de Enderezamiento/efectos de los fármacos , Convulsiones/inducido químicamente , Convulsiones/prevención & control , Estreptozocina , Tasa de Supervivencia
6.
Toxicol Ind Health ; 37(11): 695-704, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34643460

RESUMEN

Acrylonitrile (AN) is a known animal carcinogen and suspected human carcinogen. Recently, occupational exposure to AN has considerably increased. Previously, we demonstrated that streptozotocin-induced diabetes potentiates AN-induced acute toxicity in rats and that the induced cytochrome P450 2E1 (CYP2E1) is responsible for this effect. In the present study, we examined whether induction of CYP2E1 is also the underlying mechanism for the potentiation of AN-induced acute toxicity in type 2 diabetes in db/db mice. The effect of phenethyl isothiocyanate (PEITC) in reducing potentiation was also investigated. The mice were randomly divided into the normal control, diabetic control, AN, diabetes + AN, PEITC + AN, and diabetes + PEITC + AN groups. PEITC (40 mg/kg) was orally administered to rats for 3 days, and 1 h after the last PEITC gavage, 45 mg/kg AN was intraperitoneally injected. Time to death was observed. The CYP2E1 level and enzymatic activity, cytochrome c oxidase (CCO) activity, and reactive oxygen species (ROS) levels were measured. The survival rate was decreased in AN-treated db/db mice compared with that in AN-treated wild-type mice. The hepatic CYP2E1 level and enzymatic activity remained unaltered in db/db mice. Phenethyl isothiocyanate alleviated AN-induced acute toxicity in db/db mice as evident in the increased survival rate, restored CCO activity, and decreased ROS level in both the liver and brain. The study results suggested that CYP2E1 may not be responsible for the sensitivity to AN-induced acute toxicity in db/db mice and that PEITC reduced the potentiation of AN-induced acute toxicity in db/db mice.


Asunto(s)
Acrilonitrilo/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Animales , Citocromo P-450 CYP2E1/análisis , Isotiocianatos , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno , Tasa de Supervivencia
7.
Toxicol Ind Health ; 35(5): 387-397, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30991910

RESUMEN

Cytochrome P450 2E1 (CYP2E1) can be induced by diabetes mellitus, nonalcoholic liver disease, and obesity. This study assessed the protective effects of three sulfur compounds, namely phenethyl isothiocyanate (PEITC), dimethyl trisulfide (DMTS), and sodium thiosulfate (STS), on acrylonitrile (ACN)-induced acute toxicity in rats enriched with CYP2E1. PEITC and DMTS were administered intragastrically (i.g.), whereas STS was injected intraperitoneally (i.p.) at an identical dose of 0.5 mmol/kg for 3 days in acetone-pretreated rats before ACN (90 mg/kg) injection (i.p.). Acetone-treated rats that expressed high levels of CYP2E1 were more susceptible to ACN-induced acute toxicity. The sulfur compounds reduced the rate of convulsions and loss of the righting reflex in acute ACN-exposed CYP2E1-induced rats; PEITC and DMTS also increased the survival rates. PEITC inhibited hepatic CYP2E1 activity and protected hepatic and cerebral cytochrome c oxidase (CcOx) activities in acute ACN-exposed CYP2E1-enriched rats; DMTS protected hepatic CcOx activity. DMTS attenuated ACN-induced oxidative injury by reducing malondialdehyde (MDA) levels and increasing glutathione content in the brain. STS only reduced cerebral MDA levels, whereas PEITC did not exhibit any antioxidant effects. Collectively, PEITC provided superior protective effects against ACN-induced acute toxicity in rats with increased CYP2E1 activity, followed by DMTS; STS provided limited effects. PEITC and DMTS might be considered as promising chemopreventive agents against ACN-induced acute toxicity in vulnerable subpopulations with increased CYP2E1 activity.


Asunto(s)
Acrilonitrilo/toxicidad , Isotiocianatos/farmacología , Reflejo de Enderezamiento/efectos de los fármacos , Convulsiones/prevención & control , Sulfuros/farmacología , Tiosulfatos/farmacología , Animales , Citocromo P-450 CYP2E1/administración & dosificación , Sistema Enzimático del Citocromo P-450/análisis , Estimación de Kaplan-Meier , Masculino , Mortalidad , Distribución Aleatoria , Ratas , Convulsiones/inducido químicamente , Compuestos de Azufre/farmacología
8.
Arch Toxicol ; 92(6): 2093-2108, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29725710

RESUMEN

Hydrogen sulfide (H2S), the third gasotransmitter, has been shown to act as a neuroprotective factor in numerous pathological processes; however, its underlying mechanism(s) of action remain unclear. It is widely accepted that activation of moderate autophagy and the Nrf2/ARE signaling pathway play important roles in the biological self-defense systems. In the present study, we investigated whether exogenous H2S protects against the cytotoxicity of acrylonitrile (AN), a neurotoxin, in primary rat astrocytes. We found that pretreatment for 1 h with sodium hydrosulfide (NaHS), a donor of H2S (200-800 µM), significantly attenuated the AN-induced decrease in cell viability, increase in lactate dehydrogenase release and morphological changes. Furthermore, NaHS significantly attenuated AN-induced oxidative stress by reducing reactive oxygen species (ROS) levels and increasing glutathione (GSH) concentration. Moreover, NaHS activated the autophagic flux, detectable as a change in autophagy-related proteins (Beclin-1, Atg5 and p62), the formation of acidic vesicular organelles and LC3B aggregation, confirmed by adenoviral expression of mRFP-GFP-LC3. Additionally, NaHS stimulated translocation of Nrf2 into the nucleus and increased expression of heme oxygenase-1 and γ-glutamylcysteine synthetase, downstream targets of Nrf2. Notably, the autophagy inhibitor 3-methyladenine and Beclin-1, or Nrf2-targeted siRNA, significantly attenuated the neuroprotective effects of NaHS against AN-induced neurotoxicity. In conclusion, we identified a crucial role of  autophagy and the Nrf2/ARE signaling pathway in H2S-mediated neuroprotection against AN-induced toxicity in primary rat astrocytes. Our findings provide novel insights into the mechanisms of H2S-mediated neuroprotection, and suggest that H2S-based donors may serve as potential new candidate drugs to treat AN-induced neurotoxicity.


Asunto(s)
Acrilonitrilo/toxicidad , Elementos de Respuesta Antioxidante , Astrocitos/efectos de los fármacos , Autofagia/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Células Cultivadas , Cultivo Primario de Células , Ratas Sprague-Dawley , Transducción de Señal
9.
Toxicology ; 501: 153698, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38065397

RESUMEN

Dibromoacetonitrile (DBAN) is a high-risk haloacetonitrile (HAN) generated as a byproduct of chloramine disinfection in drinking water. DBAN-induced neurotoxicity in mouse hippocampal neuronal cells (HT22) and mammals was observed to be related to reactive oxygen species (ROS). ROS, endoplasmic reticulum stress (ERS) and autophagy play crucial roles in regulating a variety of cellular processes. However, whether ERS and autophagy are associated with HAN-responsive apoptosis remains unclear. This study indicated that DBAN (10 µM, 24 h) activated the ERS protein kinase like endoplasmic reticulum kinase (PERK) signaling pathway. The ERS inhibitor 4-phenylbutyric acid (4-PBA) reversed DBAN-inhibited cell viability and alleviated DBAN-induced apoptosis in HT22 cell, indicating that activation of the ERS PERK pathway mediates DBAN induced cytotoxicity. Moreover, DBAN activated autophagy. The autophagy inhibitor 3-methyladenine(3-MA) reversed DBAN-inhibited cell viability and alleviated DBAN-induced apoptosis in HT22 cell, suggesting that autophagy activation mediates DBAN-induced cell toxicity. Notably, the results showed that 4-PBA inhibited DBAN-activated autophagy, demonstrating that ERS-PERK promotes DBAN-induced cellular autophagy. Pretreatment with antioxidant N-acetylcysteine (NAC) inhibited the increase in ROS production and the activation of ERS, and protected cells from toxicity. Furthermore, 4-PBA pretreatment reduced the increase in ROS production, indicating that the ROS and PERK promote each other and form a positive feedback loop. ROS also promoted DBAN-induced autophagy. In summary, our findings indicate that DBAN induced autophagy by mediating the PERK signalling pathway and ROS interaction, leading to HT22 cell damage. Accordingly, targeting these pathogenic mechanisms may provide a potential target and theoretical basis for preventing and improving HAN-induced neurotoxicity.


Asunto(s)
Proteínas Quinasas , Transducción de Señal , Ratones , Animales , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas/metabolismo , Retículo Endoplásmico/metabolismo , Autofagia , Estrés del Retículo Endoplásmico , Apoptosis , Mamíferos/metabolismo
10.
Toxicol Res (Camb) ; 11(2): 374-384, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35510234

RESUMEN

Background: Hydrogen sulfide (H2S), as the third gasotransmitter participates in both cellular physiological and pathological processes, including chemical-induced injuries. We recently reported acute acrylonitrile (AN) treatment inhibited endogenous H2S biosynthesis pathway in rat and astrocyte models. However, there is still no evidence to address the correlation between endogenous H2S and sub-chronic AN exposure. Objectives: This study aims to explore the modulatory effects of prolonged AN exposure on endogenous H2S levels and its biosynthetic enzymes in rat blood, brain and liver. Methods: A total of 50 male Sprague-Dawley rats were randomly divided into 5 groups, including the control group and AN-treated groups at dosages of 6.25, 12.5, 25 or 50 mg/kg. Rats received one exposure/day, 5 days/week, for 4 consecutive weeks. The rat bodyweight and brain/liver organ coefficient were detected, along with liver cytochrome P450 2E1(CYP2E1) expression. In addition, the H2S contents in rat serum and plasma, and in cerebral cortex and liver tissues were measured by methylene blue method. The expression of H2S-generating enzymes, including cystathionine ß-synthase (CBS), cystathionine γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MPST) was also measured with Western blot both in rat cerebral cortex and liver. Results: Subchronic exposure to AN significantly inhibited bodyweight-gain and increased the liver CYP2E1 expression compared with the control. In addition, AN significantly increased H2S levels in rat plasma and serum, but not in liver. The endogenous H2S level in rat cerebral cortex was also significantly increased upon AN treatment, when expression of the major H2S-generating enzymes, CBS and 3-MPST were significantly enhanced. However, hepatic protein levels of CBS and CSE were significantly increased, whereas hepatic levels of 3-MPST were significantly decreased. Conclusion: This study showed that sub-chronic AN exposure increased endogenous H2S contents in rat blood and brain tissues, but not liver, which may be resulted from the distinct expression profile of H2S-producing enzymes in response to AN. The blood H2S contents may be applied as a potential novel biomarker for surveillance of chronically AN-exposed populations. Highlights: Subchronic intraperitoneal exposure to acrylonitrile increased H2S content in rat blood and cerebral cortex, but not in liver.Distinct tissue expression profiles of H2S-producing enzymes contribute to the acrylonitrile-induced differential effects on the H2S level.Blood H2S level may be a biomarker for subchronic exposure to acrylonitrile.

11.
Toxics ; 10(6)2022 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35736945

RESUMEN

Cytochrome P450 2E1 (CYP2E1) plays an essential role in the susceptibility to acute acrylonitrile (AN)-induced toxicity. Here, we investigated the toxicity and mechanism of AN in fasting mice and potential underlying mechanisms. Convulsions, loss of righting reflex, and death 4 h after AN treatment were observed and recorded for each group of mice. Relative to ad lib-fed mice, 48 h fasting significantly increased the acute toxicity of AN, as noted by a more rapid onset of convulsions and death. In addition, fasting significantly enhanced CYP2E1-mediated oxidative metabolism of AN, resulting in increased formation of CN- (one of the end-metabolites of AN). Moreover, fasting decreased hepatic GSH content, abrogating the detoxification of GSH. However, trans-1,2-dichloroethylene (DCE), a CYP2E1 inhibitor, altered the level of hepatic CYP2E1 activity in response to fasting, reduced the acute toxic symptoms of AN and the content of CN- in AN-treated mice. These data establish that fasting predisposes to AN toxicity, attributable to induced CYP2E1 and reduced hepatic GSH.

12.
Toxicol Lett ; 365: 61-73, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35724848

RESUMEN

Dibromoacetonitrile (DBAN) and dichloroacetonitrile (DCAN) are haloacetonitriles (HANs) produced as by-products of chloramine disinfection of drinking water and can cause neurotoxicity. The molecular pathways leading to HAN-induced neuronal cell death remain unclear. The nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of oxidation reactions. We explored the role of the sequestosome 1 (p62)-Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 pathway in DBAN- and DCAN-induced mouse hippocampal neuronal (HT22) cell injury. DBAN and DCAN reduced cell viability, increased lactate dehydrogenase release rate, and promoted apoptosis. Over the same treatment time, DBAN at lower concentrations caused cell injury, suggesting that DBAN is more cytotoxic than DCAN. DBAN and DCAN triggered oxidative stress by reducing intracellular glutathione and increasing reactive oxygen species concentrations. DBAN and DCAN activated the Nrf2 pathway. Furthermore, Nrf2 inhibitors (all-trans retinoic acid) attenuated DBAN- and DCAN-induced toxicity, whereas Nrf2 activators (tert-Butylhydroquinone) achieved the opposite effect. This indicates that activation of the Nrf2 pathway mediates DBAN- and DCAN-induced cell injury. Notably, the expression of p62, a noncanonical pathway that mediates Nrf2 activation, increased, whereas the expression of Keap1, another regulator of Nrf2, decreased. We noted that high p62 expression activated the Nrf2 pathway, and p62 was regulated through Nrf2, forming a positive feedback loop. N-acetyl-L-cysteine, a mercaptan substance, protected against DBAN- and DCAN-induced toxicity and inhibited the Nrf2 pathway. In summary, Nrf2 pathway inhibition and mercaptan supplementation prevent DBAN- and DCAN-induced HT22 cell injury, accordingly, targeting them is a potential approach to preventing HAN-induced neurotoxicity.


Asunto(s)
Agua Potable , Acetonitrilos , Acetilcisteína/farmacología , Animales , Desinfección , Hipocampo , Proteína 1 Asociada A ECH Tipo Kelch , Ratones , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Compuestos de Sulfhidrilo
13.
Neurotoxicology ; 90: 228-236, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35439497

RESUMEN

OBJECTIVE: Ferroptosis is an iron-dependent nonapoptotic form of cell death, characterized by iron accumulation and lipid peroxidation. However, the role of ferroptosis in methylmercury (MeHg)-induced cytotoxicity has yet to be fully characterized. The purpose of this study was to investigate the role of ferroptosis in MeHg-induced cytotoxicity in both brain and liver cells. METHODS: The effects of MeHg on cell viability, cytotoxicity, intracellular iron content, reduced glutathione (GSH) content, ferroptosis-related proteins, cytosolic and lipid reactive oxygen species (ROS) generation were determined in rat primary astrocytes (AST) and Buffalo Rat Liver (BRL) cells in the absence or presence of the ferroptosis inhibitors deferoxamine (DFO) or ferrostatin-1 (Fer-1). RESULTS: MeHg treatment decreased cell viability and increased cytotoxicity in AST and BRL cells. MeHg induced ferroptosis in AST and BRL cells was reflected by increased cytosolic ROS, lipid ROS and intracellular iron content, all of which were inhibited by the ferroptosis inhibitors DFO and/or Fer-1. MeHg inhibited the expression of ferritin heavy chain 1 (FTH1). Furthermore, MeHg treatment decreased the expression of glutathione peroxidase 4 (GPx4) without altering solute carrier family 7 member 11 (SLC7A11). DFO and Fer-1 significantly increased the expression of GPx4, yet had no effect on SLC7A11 upon MeHg treatment. CONCLUSIONS: Our novel results are consistent with ferroptosis as a key event mediating MeHg-induced toxicity, inhibiting GPx4 in AST and BRL cells. Ferroptosis may offer a new target for attenuating MeHg-induced toxic injury.


Asunto(s)
Ferroptosis , Compuestos de Metilmercurio , Animales , Astrocitos/metabolismo , Hierro/metabolismo , Lípidos , Hígado/metabolismo , Compuestos de Metilmercurio/toxicidad , Ratas , Especies Reactivas de Oxígeno/metabolismo
14.
Toxicology ; 451: 152685, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33486070

RESUMEN

Hydrogen sulfide (H2S) as the third gasotransmitter molecule serves various biological regulatory roles in health and disease. Acrylonitrile (AN) is a common occupational toxicant and environmental pollutant, causing brain and liver damage in mammals. The biotransformation of AN is dependent-upon reduced glutathione (GSH), cysteine and other sulfur-containing compounds. However, the effects of AN on the endogenous H2S biosynthesis pathway have yet to be determined. Herein, we demonstrated that a single exposure to AN (at 25, 50, or 75 mg/kg for 1, 6 or 24 h) decreased the endogenous H2S content and H2S-producing capacity in a dose-dependent manner, both in the cerebral cortex and liver of rats in vivo. In addition, the inhibitory effects of AN (1, 2.5, 5, 10 mM for 12 h) on the H2S content and/or the expression of H2S-producing enzymes were also found both in primary rat astrocytes and rat liver cell line (BRL cells). Impairment in the H2S biosynthesis pathway was also assessed in primary rat astrocytes treated with AN. It was found that inhibition of the cystathionine-ß-synthase (CBS)/3-mercaptopyruvate sulfurtransferase (3-MPST)-H2S pathway with the CBS inhibitor or 3-MPST-targeted siRNA significantly increased the AN-induced (5 mM for 12 h) cytotoxicity in astrocytes. In turn, CBS activation or 3-MPST overexpression as well as exogenous NaHS supplementation significantly attenuated AN-induced cytotoxicity. Taken together, endogenous H2S biosynthesis pathway was disrupted in rats acutely exposed to AN, which contributes to acute AN neurotoxicity in primary rat astrocytes.


Asunto(s)
Acrilonitrilo/toxicidad , Astrocitos/metabolismo , Encéfalo/metabolismo , Cistationina betasintasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Hígado/metabolismo , Sulfurtransferasas/metabolismo , Animales , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sulfuro de Hidrógeno/antagonistas & inhibidores , Hígado/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
Wei Sheng Yan Jiu ; 39(3): 271-4, 2010 May.
Artículo en Zh | MEDLINE | ID: mdl-20568450

RESUMEN

OBJECTIVE: To explore time-course effect and region-specificity of endoplasmic reticulum stress in rat brain acutely exposed by methylmercury (MeHg). METHODS: Forty-two SD rats were randomly divided into seven groups, and the rats intraperitoneally injected at the dose of 4 mg/kg bw. MeHg were decapitated at the times of 0.5, 1, 3, 6, 12 and 24 h, then rats in control group intraperitoneally injected by the corresponding volume of normal saline were decapitated. The cerebellum, cerebral cortex, brain stem, hippocampus and striatum were dissected out and weighted on ice at once. The expressions of Grp78 protein which is a marker of endoplasmic reticulum stress, were determined by Western blotting analysis. The contents of reduced glutathione (GSH) of cerebral cortex were also determined. RESULTS: After exposure to MeHg, the tendencies of expression of Grp78 were consistent in various brain regions. It began to increase at the time of 0.5 h and the peak levels reached at the times of 6 h or 12 h, then it began to decline. All expression levels returned nearly to the control levels at the time of 24h. The alternations of Grp78 protein in cerebral cortex and brain stem were statistically significant in comparison with those of control groups at the time of 0h among brain regions. Increases of Grp78 protein in cerebral cortex reached peak levels at the time of 6h after MeHg exposure, and the expressions of Grp78 protein corresponded to 150% of control levels. Increases of Grp78 protein in brain stem reached peak levels at the time of 12 h after MeHg exposure, and the expressions of Grp78 protein corresponded to 140% of control levels. Further, the contents of GSH in cerebral cortex showed the tendencies of first decreases and then gradually showed increases. These changes were inversely correlated to the change of Grp78 protein in cerebral cortex (r = -0.77). CONCLUSION: Rats acutely exposed with MeHg could show endoplasmic reticulum stress in a time dependent and region-specific pattern, and this alteration could be associated with oxidative stress in cerebral cortex.


Asunto(s)
Encéfalo/metabolismo , Estrés del Retículo Endoplásmico , Exposición a Riesgos Ambientales/efectos adversos , Compuestos de Metilmercurio/toxicidad , Estrés Oxidativo/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
16.
Toxicol Lett ; 331: 82-91, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32461003

RESUMEN

Hypoxia-inducible factor 1 (HIF-1) is a critical nuclear transcription factor for adaptation to hypoxia; its regulatable subunit, HIF-1α, is a cytoprotective regulatory factor. We examined the effects of methylmercury (MeHg) in rat adrenal pheochromocytoma (PC12) cells and the rat hepatocyte cell line BRL. MeHg treatment led to time- and concentration-dependent toxicity in both lines with statistically significant cytotoxic effects at 5 µM and 10 µM in PC12 and BRL, respectively, at 0.5 h. HIF-1α protein levels were significantly decreased at 2.5 (PC12) and 5 (BRL) µM MeHg. Furthermore, MeHg reduced the protein levels of HIF-1α and its target genes (glucose transporter-1, vascular endothelial growth factor-A and erythropoietin). Overexpression of HIF-1α significantly attenuated MeHg-induced toxicity in both cell types. Notably, cobalt chloride, a pharmacological inducer of HIF-1α, significantly attenuated MeHg-induced toxicity in BRL but not PC12. In both cell lines, an inhibitor of prolyl hydroxylase, 3, 4-dihydroxybenzoic acid, and the proteasome inhibitor carbobenzoxy-L-leucyl-L-leucyl-L-leucinal(MG132), antagonized MeHg toxicity, while 2-methoxyestradiol, a HIF-1α inhibitor, significantly increased it. These data establish that: (a) neuron-like PC12 cells are more sensitive to MeHg than non-neuronal BRL cells; (b) HIF-1α plays a similar role in MeHg-induced toxicity in both cell lines; and (c) upregulation of HIF-1α offers general cytoprotection against MeHg toxicity in PC12 and BRL cell lines.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Compuestos de Metilmercurio/toxicidad , Neuronas/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células PC12 , ARN Mensajero/metabolismo , Ratas , Transducción de Señal , Regulación hacia Arriba
17.
Genes (Basel) ; 10(2)2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30754717

RESUMEN

The WRKY gene family plays a unique role in plant stress tolerance. Quinoa is a cultivated crop worldwide that is known for its high stress tolerance. The WRKY gene family in quinoa has not yet been studied. Using a genome-wide search method, we identified 1226 WRKY genes in 15 plant species, seven animal species, and seven fungi species. WRKY proteins were not found in animal species and five fungi species, but were, however, widespread in land plants. A total of 92 CqWRKY genes were identified in quinoa. Based on the phylogenetic analysis, these CqWRKY genes were classified into three groups. The CqWRKY proteins have a highly conserved heptapeptide WRKYGQK with 15 conserved elements. Furthermore, a total of 25 CqWRKY genes were involved in the co-expression pathway of organ development and osmotic stress. The expression level of more than half of these CqWRKY genes showed significant variation under salt or drought stress. This study reports, for the first time, the findings of the CqWRKY gene family in quinoa at the genome-wide level. This information will be beneficial for our understanding of the molecular mechanisms of stress tolerance in crops, such as quinoa.


Asunto(s)
Chenopodium quinoa/genética , Evolución Molecular , Proteínas de Plantas/genética , Factores de Transcripción/genética , Secuencias de Aminoácidos , Chenopodium quinoa/clasificación , Chenopodium quinoa/metabolismo , Secuencia Conservada , Regulación de la Expresión Génica de las Plantas , Familia de Multigenes , Presión Osmótica , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Estrés Salino , Factores de Transcripción/química , Factores de Transcripción/metabolismo
18.
Toxicology ; 425: 152248, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31330227

RESUMEN

Methylmercury (MeHg) is a ubiquitous environmental toxicant that leads to long-lasting neurological deficits in animals and humans. Curcumin, a polyphenol obtained from the rhizome of turmeric, has well-known antioxidant functions. Here, we evaluated curcumin's efficacy in mitigating MeHg-induced cytotoxicity and further investigated the underlying mechanism of this neuroprotection in primary rat astrocytes. Pretreatment with curcumin (2, 5, 10 and 20 µM for 3, 6, 12 or 24 h) protected against MeHg-induced (5 µM for 6 h) cell death in a time and dose-dependent manner. Curcumin (2, 5, 10 or 20 µM) pretreatment for 12 h significantly ameliorated the MeHg-induced astrocyte injury and oxidative stress, as evidenced by morphological alterations, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) generation, and glutathione (GSH) and catalase (CAT) levels. Moreover, curcumin pretreatment increased Nrf2 nuclear translocation and downstream enzyme expression, heme oxygenase-1 (HO-1) and NADPH quinone reductase-1 (NQO1). Knockdown of Nrf2 with siRNA attenuated the protective effect of curcumin against MeHg-induced cell death. However, both the pan-protein kinase C (PKC) inhibitor, Ro 31-8220, and the selective PKCδ inhibitor, rottlerin, failed to suppress the curcumin-activated Nrf2/Antioxidant Response Element(ARE) pathway and attenuate the protection exerted by curcumin. Taken together, these findings confirm that curcumin protects against MeHg-induced neurotoxicity by activating the Nrf2/ARE pathway and this protection is independent of PKCδ activation. More studies are needed to understand the mechanisms of curcumin cytoprotection.


Asunto(s)
Elementos de Respuesta Antioxidante/genética , Astrocitos/efectos de los fármacos , Curcumina/farmacología , Compuestos de Metilmercurio/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Acetofenonas/farmacología , Animales , Benzopiranos/farmacología , Relación Dosis-Respuesta a Droga , Técnica del Anticuerpo Fluorescente , Glutatión/metabolismo , Hylobatidae , Indoles/farmacología , L-Lactato Deshidrogenasa/metabolismo , Compuestos de Metilmercurio/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Proteína Quinasa C-delta/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
19.
Environ Health Perspect ; 127(12): 127006, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31850806

RESUMEN

BACKGROUND: As a ubiquitous environmental pollutant, methylmercury (MeHg) induces toxic effects in the nervous system, one of its main targets. However, the exact mechanisms of its neurotoxicity have not been fully elucidated. Hypoxia-inducible factor-1α (HIF-1α), a transcription factor, plays a crucial role in adaptive and cytoprotective responses in cells and is involved in cell survival, proliferation, apoptosis, inflammation, angiogenesis, glucose metabolism, erythropoiesis, and other physiological activities. OBJECTIVES: The aim of this study was to explore the role of HIF-1α in response to acute MeHg exposure in rat brain and primary cultured astrocytes to improve understanding of the mechanisms of MeHg-induced neurotoxicity and the development of effective neuroprotective strategies. METHODS: Primary rat astrocytes were treated with MeHg (0-10µM) for 0.5h. Cell proliferation and cytotoxicity were assessed with a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl diphenyltetrazolium bromide (MTT) assay and a lactate dehydrogenase (LDH) release assay, respectively. Reactive oxygen species (ROS) levels were analyzed to assess the level of oxidative stress using 2',7'-dichlorofluorescin diacetate (DCFH-DA) fluorescence. HIF-1α, and its downstream proteins, glucose transporter 1 (GLUT-1), erythropoietin (EPO), and vascular endothelial growth factor A (VEGF-A) were analyzed by means of Western blotting. Real-time PCR was used to detect the expression of HIF-1α mRNA. Pretreatment with protein synthesis inhibitor (CHX), proteasome inhibitor (MG132), or proline hydroxylase inhibitor (DHB) were applied to explore the possible mechanisms of HIF-1α inhibition by MeHg. To investigate the role of HIF-1α in MeHg-induced neurotoxicity, cobalt chloride (CoCl2), 2-methoxyestradiol (2-MeOE2), small interfering RNA (siRNA) transfection and adenovirus overexpression were used. Pretreatment with N-acetyl-L-cysteine (NAC) and vitamin E (Trolox) were used to investigate the putative role of oxidative stress in MeHg-induced alterations in HIF-1α levels. The expression of HIF-1α and related downstream proteins was detected in adult rat brain exposed to MeHg (0-10mg/kg) for 0.5h in vivo. RESULTS: MeHg caused lower cell proliferation and higher cytotoxicity in primary rat astrocytes in a time- and concentration-dependent manner. In comparison with the control cells, exposure to 10µM MeHg for 0.5h significantly inhibited the expression of astrocytic HIF-1α, and the downstream genes GLUT-1, EPO, and VEGF-A (p<0.05), in the absence of a significant decrease in HIF-1α mRNA levels. When protein synthesis was inhibited by CHX, MeHg promoted the degradation rate of HIF-1α. MG132 and DHB significantly blocked the MeHg-induced decrease in HIF-1α expression (p<0.05). Overexpression of HIF-1α significantly attenuated the decline in MeHg-induced cell proliferation, whereas the inhibition of HIF-1α significantly increased the decline in cell proliferation (p<0.05). NAC and Trolox, two established antioxidants, reversed the MeHg-induced decline in HIF-1α protein levels and the decrease in cell proliferation (p<0.05). MeHg suppressed the expression of HIF-1α and related downstream target proteins in adult rat brain. DISCUSSION: MeHg induced a significant reduction in HIF-1α protein by activating proline hydroxylase (PHD) and the ubiquitin proteasome system (UPS) in primary rat astrocytes. Additionally, ROS scavenging by antioxidants played a neuroprotective role via increasing HIF-1α expression in response to MeHg toxicity. Moreover, we established that up-regulation of HIF-1α might serve to mitigate the acute toxicity of MeHg in astrocytes, affording a novel therapeutic target for future exploration. https://doi.org/10.1289/EHP5139.


Asunto(s)
Contaminantes Ambientales/toxicidad , Compuestos de Metilmercurio/toxicidad , Pruebas de Toxicidad Aguda , Animales , Astrocitos , Encéfalo , Proliferación Celular , Supervivencia Celular , Regulación de la Expresión Génica , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , ARN Mensajero/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular
20.
Genes (Basel) ; 9(2)2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29389910

RESUMEN

The homeodomain-leucine zipper (HD-Zip) gene family, as plant-specific transcription factors, plays an important role in plant development and growth as well as in the response to diverse stresses. Although HD-Zip genes have been extensively studied in many plants, they had not yet been studied in wheat, especially those involved in response to abiotic stresses. In this study, 46 wheat HD-Zip genes were identified using a genome-wide search method. Phylogenetic analysis classified these genes into four groups, numbered 4, 5, 17 and 20 respectively. In total, only three genes with A, B and D homoeologous copies were identified. Furthermore, the gene interaction networks found that the TaHDZ genes played a critical role in the regulatory pathway of organ development and osmotic stress. Finally, the expression profiles of the wheat HD-Zips in different tissues and under various abiotic stresses were investigated using the available RNA sequencing (RNA-Seq) data and then validated by quantitative real-time polymerase chain reaction (qRT-PCR) to obtain the tissue-specific and stress-responsive candidates. This study systematically identifies the HD-Zip gene family in wheat at the genome-wide level, providing important candidates for further functional analysis and contributing to the better understanding of the molecular basis of development and stress tolerance in wheat.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA