Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 758
Filtrar
1.
Cardiovasc Toxicol ; 24(6): 539-549, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703273

RESUMEN

NaAsO2 is known as a harmful pollutant all over the world, and many chronic heart diseases can be attributed to its prolonged exposure in NaAsO2-contaminated water. Therefore, considering the anti-inflammatory and antioxidant effects of betaine (BET), in this study, our team investigated the cardioprotective effects of this phytochemical agent on sodium arsenite (NaAsO2)-induced cardiotoxicity. Forty male mice were randomly divided into 4 groups: (I) Control; (II) BET (500 mg/kg); (III) NaAsO2 (50 ppm); and (IV) NaAsO2 + BET. NaAsO2 was given to the animals for 8 weeks, but BET was given in the last two weeks. After decapitation, inflammatory factors and biochemical parameters were measured, and Western blot analyses were performed. BET decrease the activity level of alanine aspartate aminotransferase, creatine kinase MB, thiobarbituric acid reactive substances level, inflammatory factors (tumor necrosis factor-α) content, and nuclear factor kappa B expression. Furthermore, BET increased cardiac total thiol and activity levels of catalase, superoxide dismutase, and glutathione peroxidase and nuclear factor erythroid-2 expression. Hence, the administration of BET ameliorated the deleterious effects stemming from the imbalance of oxidative and antioxidant pathways and histopathological alterations observed in NaAsO2-intoxicated mice, thereby attenuating oxidative stress-induced damage and inflammation.


Asunto(s)
Antiinflamatorios , Antioxidantes , Arsenitos , Betaína , Cardiotoxicidad , Modelos Animales de Enfermedad , Cardiopatías , Mediadores de Inflamación , Estrés Oxidativo , Transducción de Señal , Compuestos de Sodio , Animales , Arsenitos/toxicidad , Compuestos de Sodio/toxicidad , Masculino , Antioxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Antiinflamatorios/farmacología , Ratones , Betaína/farmacología , Cardiopatías/prevención & control , Cardiopatías/inducido químicamente , Cardiopatías/patología , Cardiopatías/metabolismo , Mediadores de Inflamación/metabolismo , Transducción de Señal/efectos de los fármacos , Biomarcadores/metabolismo , Biomarcadores/sangre , Citoprotección , Miocardio/patología , Miocardio/metabolismo
2.
Toxicology ; 504: 153795, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574842

RESUMEN

The mechanistic target of rapamycin (RAPA) complex 1 (mTORC1) - transcription factor EB (TFEB) pathway plays a crucial role in response to nutritional status, energy and environmental stress for maintaining cellular homeostasis. But there is few reports on its role in the toxic effects of arsenic exposure and the related mechanisms. Here, we show that the exposure of bronchial epithelial cells (BEAS-2B) to sodium arsenite promoted the activation of mTORC1 (p-mTORC1) and the inactivation of TFEB (p-TFEB), the number and activity of lysosomes decreased, the content of reduced glutathione (GSH) and superoxide dismutase (SOD) decreased, the content of malondialdehyde (MDA) increased, the DNA and chromosome damage elevated. Further, when mTORC1 was inhibited with RAPA, p-mTORC1 and p-TFEB down-regulated, GSH and SOD increased, MDA decreased, the DNA and chromosome damage reduced significantly, as compared with the control group. Our data revealed for the first time that mTORC1 - TFEB pathway was involved in sodium arsenite induced lysosomal alteration, oxidative stress and genetic damage in BEAS-2B cells, and it may be a potential intervention target for the toxic effects of arsenic.


Asunto(s)
Arsenitos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Daño del ADN , Lisosomas , Diana Mecanicista del Complejo 1 de la Rapamicina , Estrés Oxidativo , Compuestos de Sodio , Arsenitos/toxicidad , Compuestos de Sodio/toxicidad , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Línea Celular , Daño del ADN/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transducción de Señal/efectos de los fármacos , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Bronquios/citología , Bronquios/patología , Glutatión/metabolismo , Superóxido Dismutasa/metabolismo , Complejos Multiproteicos/metabolismo , Malondialdehído/metabolismo
3.
Environ Toxicol ; 39(6): 3563-3577, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38477077

RESUMEN

Lysine specific demethylase 1 (LSD1) is a histone demethylase that specifically catalyzes the demethylation of histone H3K4 (H3K4me1/2) and regulates gene expression. In addition, it can mediate the process of autophagy through its demethylase activity. Sestrin2 (SESN2) is a stress-induced protein and a positive regulator of autophagy. In NaAsO2-induced mouse fibrotic livers and activated hepatic stellate cells (HSCs), LSD1 expression is decreased, SESN2 expression is increased, and autophagy levels are also increased. Overexpression of LSD1 and silencing of SESN2 decreased the level of autophagy and attenuated the activation of HSCs induced by NaAsO2. LSD1 promoted SESN2 gene transcription by increasing H3K4me1/2 in the SESN2 promoter region. 3-methyladenine (3-MA) and chloroquine were used to inhibit autophagy of HSCs, and the degree of activation was also alleviated. Taken together, LSD1 positively regulates SESN2 by increasing H3K4me1/2 enrichment in the SESN2 promoter region, which in turn increases the level of autophagy and promotes the activation of HSCs. Our results may provide new evidence for the importance of LSD1 in the process of autophagy and activation of HSCs induced by arsenic poisoning. Increasing the expression and activity of LSD1 is expected to be an effective way to reverse the autophagy and activation of HSCs induced by arsenic poisoning.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Arsenitos , Histona Demetilasas , Transducción de Señal , Compuestos de Sodio , Animales , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Transducción de Señal/efectos de los fármacos , Arsenitos/toxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Ratones , Compuestos de Sodio/toxicidad , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Autofagia/efectos de los fármacos , Masculino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones Endogámicos C57BL
4.
Toxicology ; 492: 153529, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37120063

RESUMEN

Chronic arsenic exposure causes myocardial damage. The aim of this study is to investigate if oxidative stress and reduction in NO is involved in the myocardial damage induced by arsenic in drinking water. Rats were divided into a control group and different doses of sodium arsenite. With increasing sodium arsenite concentrations in drinking water, localised inflammatory foci and necrotic myocardial tissues were gradually observed. Compared to the control group, the activities and gene expression of antioxidant enzymes in arsenic-exposed rats decreased. NO content and the NOS activity as well as the expression of NOS mRNA in the myocardial tissue of exposed rats, decreased, and the extracellular NO content of cardiomyocytes treated with sodium arsenite also decreased. The rate of cell apoptosis induced by sodium arsenite decreased after treatment with sodium nitroprusside (an NO donor). In conclusion, arsenic exposure in drinking water can lead to myocardial injury and cardiomyocyte apoptosis through oxidative stress and a reduction in NO content.


Asunto(s)
Arsénico , Arsenitos , Agua Potable , Ratas , Animales , Arsénico/toxicidad , Estrés Oxidativo , Arsenitos/toxicidad , Compuestos de Sodio/toxicidad
5.
Niger J Physiol Sci ; 37(1): 83-92, 2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35947839

RESUMEN

Arsenic compromises the gastrointestinal integrity and function via the body's anti-oxidative system breakdown.  Hence, this study aimed to investigate the effects of tocopherol on redox imbalance and histoarchitectural alterations in rats' gastrointestinal tract exposed to sodium arsenite. Sodium arsenite and graded doses of tocopherol were administered orally into experimental rats assigned to different groups for four weeks concurrently. Redox status assay was done in homogenized samples by spectrophotometry. Parietal cell mass and mucous cell density (stomach), villus height and crypt depth (ileum), goblet cells count, and crypt depth (colon) were evaluated by histomorphometry. Inflammatory cells infiltration was also assessed using a semi-quantitative procedure. Sodium arsenite caused a significant increase in Malondialdehyde and Myeloperoxidase but, decreased Superoxide dismutase, Catalase, Nitric oxide, Glutathione peroxidase, Glutathione, and Glutathione-S-Transferase. Tocopherol treatment reversed the changes (p<0.05) though not largely dose-dependent. Furthermore, tocopherol annulled sodium arsenite-induced increase in parietal cell mass and decrease in mucous cell density in the stomach, decrease in villus height and villus height/crypt depth ratio in the ileum, and decrease in goblets cells and increase in crypt depth in the colon. Moreover, activated inflammatory cell infiltration by sodium arsenite was mitigated by tocopherol. Sodium arsenite provokes not only marked inflammatory cellular infiltration but a focal loss of glands, hyperplasia of crypts, atrophic villi, and hypertrophy of Peyer's patches in the intestines, which are all lessened with tocopherol treatment.  These findings underscore the anti-oxidative properties of tocopherol as a potent dietary factor against sodium arsenite toxicity in the gastrointestinal tract. Keywords: Tocopherol, arsenic, stomach, ileum, colon.


Asunto(s)
Arsénico , Arsenitos , Animales , Antioxidantes/uso terapéutico , Arsénico/metabolismo , Arsénico/farmacología , Arsenitos/toxicidad , Tracto Gastrointestinal , Glutatión/metabolismo , Estrés Oxidativo , Ratas , Compuestos de Sodio/toxicidad , Superóxido Dismutasa/metabolismo , Tocoferoles/metabolismo , Tocoferoles/farmacología , Vitamina E/farmacología
6.
Toxicology ; 474: 153212, 2022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-35598691

RESUMEN

There is epidemiologic evidence to suggest that arsenic exposure is associated with risk of prostate cancer incidence and mortality. There are no studies indicating that arsenic can induce prostate cancer in animals. We evaluated whether drinking water exposure to sodium arsenite would affect prostate carcinogenesis in a rat model that depends on long-term low dose treatment with testosterone. WU rats received a sequential treatment with the antiandrogen flutamide followed by a single androgen administration to stimulate prostatic cell proliferation during which a single injection of N-methyl-N-nitrosourea was given. Two weeks later the animals received subcutaneous slow release testosterone-containing silastic tubing implants and provided with drinking water containing 5 mg/L sodium arsenite or with control drinking water for the duration of the 64 week-long experiment. Arsenite provided in drinking water did not modify the induction of prostate cancer in this rat model compared to control rats or survival. It also did not affect the growth of the animals or their drinking water intake. This animal study with drinking water exposure to 5 mg/L sodium arsenite does not support the notion that arsenic enhances prostate carcinogenesis.


Asunto(s)
Arsénico , Arsenitos , Agua Potable , Neoplasias de la Próstata , Animales , Arsénico/toxicidad , Arsenitos/toxicidad , Carcinogénesis/inducido químicamente , Humanos , Masculino , Próstata , Neoplasias de la Próstata/inducido químicamente , Ratas , Compuestos de Sodio/toxicidad , Testosterona
7.
Andrologia ; 54(3): e14347, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34897760

RESUMEN

The current study investigated the protective ameliorative effect of intraperitoneally administered kisspeptin-10 (50 nmol/day) against reproductive toxicity in adult male mice challenged with 35 days of exposure to sodium arsenite in drinking water. Mice were divided into tap water control, sodium arsenite-alone (4 ppm and 10 ppm), kisspeptin-alone (intermittent and continuous) and combined (sodium arsenite +kisspeptin-10 intermittent and continuous) treatment groups. Results revealed protective effect of both intermittent and continuous kisspeptin doses on reproductive organs against sodium arsenite-induced toxicity. This was indicated by an increase (p < 0.001) in the activity of antioxidant enzymes and a decrease (p < 0.001) in the levels of oxidative stress biomarkers. Concomitant significant increase was noticeable in the relative organ weight (p < 0.01), and serum testosterone and seminal fructose (p < 0.001), and a significant improvement in sperm parameters was also observed. A significant downregulation of lactate dehydrogenase concentration demonstrated further the protective effect of kisspeptin against tissue damage. Histologically, both treatment regimens of kisspeptin combined with sodium arsenite exposure prevented massive germ cell loss and tissue damage, a condition prominent in sodium arsenite-alone-treated mice. The study demonstrates for the first time kisspeptin's potential to mitigate the biochemical and histotoxic effects of arsenic on male reproductive system.


Asunto(s)
Arsenitos , Kisspeptinas , Animales , Arsenitos/toxicidad , Kisspeptinas/farmacología , Masculino , Ratones , Estrés Oxidativo , Compuestos de Sodio/toxicidad
8.
Biochem Biophys Res Commun ; 587: 16-23, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34861471

RESUMEN

Arsenic is a potent carcinogen in humans. However, the molecular mechanisms underlying its toxicity in lung cancer remain unclear. Here, we report that arsenite-induced cytotoxicity is regulated by SQSTM1/p62 and BNIP3L/Nix signaling in non-small-cell lung cancer H460 cells. Arsenite exposure resulted in dose-dependent growth inhibition, which was associated with apoptosis, as demonstrated by depolarized mitochondrial membrane potential and cleavage of caspase-8, caspase-3, PARP-1, and Bax. The autophagy adaptor p62 was detected in the monomeric and multiple high-molecular-weight (HMW) forms, and protein levels were upregulated depending on both arsenite concentrations (≤45 µM) and exposure times (<24 h). LC3-II, an autophagy marker, was upregulated as early as 1 h after arsenite treatment. Expression of Nix, a mitochondrial outer membrane protein, continued to increase with arsenite concentration and exposure time; it was detected in the monomeric and multiple HMW forms. Soon after arsenite exposure, p62 colocalized with Nix in the cytoplasm, and p62 knockdown reduced the Nix levels and increased the LC3-II levels. In contrast, Nix knockdown did not affect the p62 and LC3-II levels but reduced caspase-8, caspase-3, and Bax cleavage, indicating that Nix accumulation resulted from its reduced autophagic degradation and promoted apoptosis. p38 inhibition markedly increased arsenite-induced Nix protein and reduced p62 protein levels, resulting in increased autophagy and apoptosis. Furthermore, c-Jun NH2-terminal kinase inhibition reduced Nix and Bax cleavage, and both signaling pathways were suppressed by N-acetylcysteine treatment. Our results suggest that arsenite-induced cytotoxicity is modulated by the coordinated action of p62 and Nix through MAPK.


Asunto(s)
Arsenitos/toxicidad , Células Epiteliales/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Proteína Sequestosoma-1/genética , Compuestos de Sodio/toxicidad , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Food Chem Toxicol ; 160: 112771, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34920032

RESUMEN

As an environmental toxicant, arsenic exposure may cause insulin resistance (IR). Previous studies have shown that pyroptosis plays an important role in the occurrence and development of IR. Although gasdermin D (GSDMD) functions as an executor of pyroptosis, the relationship between GSDMD-mediated pyroptosis and hepatic IR remains unclear. Here, we observed that sodium arsenite (NaAsO2) activated NOD-like receptors containing pyrin domain 3 (NLRP3) inflammasomes, promoted GSDMD activation, induced pyroptosis and hepatic IR, while GSDMD knockdown attenuated pyroptosis and hepatic IR caused by NaAsO2. However, GSDMD interference did not affect NLRP3 activation. Ubiquitination modification is widely involved in protein regulation and intracellular signal transduction, and whether it regulates GSDMD and affects its degradation, and exerts effects on arsenic-induced pyroptosis remain unclear. We observed that NaAsO2 reduced the K48- and K63-linked ubiquitination of GSDMD, thereby inhibiting its degradation through the ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway (ALP), causing GSDMD to accumulate and lyse into GSDMD-N, which promoted pyroptosis. In summary, we demonstrated that GSDMD participated in arsenic-induced hepatic IR. Moreover, NaAsO2 reduced GSDMD ubiquitination and decreased its intracellular degradation, aggravating pyroptosis and hepatic IR. We have revealed the molecular mechanism underpinning arsenic-induced IR, and we provide potential solutions for the prevention and treatment of type 2 diabetes (T2D).


Asunto(s)
Arsenitos/toxicidad , Resistencia a la Insulina , Hígado/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Piroptosis/efectos de los fármacos , Compuestos de Sodio/toxicidad , Animales , Línea Celular , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Ratas , Ratas Sprague-Dawley , Ubiquitinación/efectos de los fármacos
10.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-34830013

RESUMEN

Mercury ranks third on the U.S. Agency of Toxic Substances and Disease Registry priority list of hazardous substances, behind only arsenic and lead. We have undertaken uncovering the mechanisms underlying the developmental toxicity of methylmercury (MeHg), inorganic mercury (HgCl2), lead acetate (Pb), and sodium arsenite (As). To probe these differences, we used the Drosophila model, taking advantage of three developmental transitions-pupariation, metamorphosis, and eclosion-to differentiate potentially unique windows of toxicity. We elaborated dose response profiles for each individual metal administered in food and accounted for internal body burden, also extending analyses to evaluate combinatorial metal mixture effects. We observed all four metals producing larval lethality and delayed pupariation, with MeHg being most potent. Compared to other metals, MeHg's potency is caused by a higher body burden with respect to dose. MeHg uniquely caused dose-dependent failure in eclosion that was unexpectedly rescued by titrating in HgCl2. Our results highlight a unique developmental window and toxicokinetic properties where MeHg acts with specificity relative to HgCl2, Pb, and As. These findings will serve to refine future studies aimed at revealing tissue morphogenesis events and cell signaling pathways, potentially conserved in higher organisms, that selectively mediate MeHg toxicity and its antagonism by HgCl2.


Asunto(s)
Drosophila melanogaster/efectos de los fármacos , Mercurio/toxicidad , Metales/toxicidad , Compuestos de Metilmercurio/toxicidad , Animales , Arsenitos/toxicidad , Drosophila melanogaster/crecimiento & desarrollo , Humanos , Larva/efectos de los fármacos , Compuestos Organometálicos/toxicidad , Isoformas de Proteínas/toxicidad , Compuestos de Sodio/toxicidad , Fenómenos Toxicológicos
11.
Tissue Cell ; 73: 101657, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34628213

RESUMEN

BACKGROUND: We aimed to study the beneficial property of chrysin (CHR) by targeting its antioxidant and anti-inflammatory effects on nephrotoxicity induced by sodium arsenite (SA). MATERIALS & METHODS: We have used the 35 male Wistar rats in five equal groups (n = 7). Normal saline in (5 ml/kg; p.o.; 21 days) was given to the control group. Sodium arsenite (10 mg/kg; p.o.; 14 days) was given to the SA group. CHR (25, 50 and 100 mg/kg; p.o.; 21 days) and SA (10 mg/kg; p.o.; 14 days from the 7th day of the experiment) was given to the SA + CHR 25, 50 and 100 groups. On the 22nd day of the experiment, the animals' bloods and kidneys were taken, and then we have performed functional, biochemical and histological assessment. RESULTS: CHR pre- and alongside administration (more potently at dose of 100 mg/kg) with SA reduced the SA-induced alterations in serum creatinine and blood urine nitrogen levels. Increased levels of protein carbonyl, myeloperoxidase, malondialdehyde and nitric oxide in kidney tissue were decreased by CHR treatment. CHR administration increased the levels of glutathione and activities of glutathione peroxidase, catalase and superoxide dismutase in renal tissue. Moreover, treatment with CHR reduced the levels of inflammatory mediators including interleukin 1 beta and tumor necrosis factor alpha in renal tissue. The renal histological lesions induced SA were mitigated by CHR treatment in dose dependent manner. CONCLUSION: The results of present study suggested that administration of CHR before and alongside with SA attenuated the renal toxic effects of SA via antioxidative stress and anti-inflammatory effects.


Asunto(s)
Arsenitos/toxicidad , Flavonoides/farmacología , Inflamación/patología , Riñón/patología , Estrés Oxidativo , Compuestos de Sodio/toxicidad , Animales , Antioxidantes/metabolismo , Catalasa/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Malondialdehído/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Peroxidasa/metabolismo , Carbonilación Proteica/efectos de los fármacos , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
12.
Nutrients ; 13(8)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34445052

RESUMEN

Chronic arsenic exposure via drinking water is associated with diabetes in human pop-ulations throughout the world. Arsenic is believed to exert its diabetogenic effects via multiple mechanisms, including alterations to insulin secretion and insulin sensitivity. In the past, acute arsenicosis has been thought to be partially treatable with selenium supplementation, though a potential interaction between selenium and arsenic had not been evaluated under longer-term exposure models. The purpose of the present study was to explore whether selenium status may augment arsenic's effects during chronic arsenic exposure. To test this possibility, mice were exposed to arsenic in their drinking water and provided ad libitum access to either a diet replete with selenium (Control) or deficient in selenium (SelD). Arsenic significantly improved glucose tolerance and decreased insulin secretion and ß-cell function in vivo. Dietary selenium deficiency resulted in similar effects on glucose tolerance and insulin secretion, with significant interactions between arsenic and dietary conditions in select insulin-related parameters. The findings of this study highlight the complexity of arsenic's metabolic effects and suggest that selenium deficiency may interact with arsenic exposure on ß-cell-related physiological parameters.


Asunto(s)
Arsenitos/toxicidad , Glucemia/efectos de los fármacos , Enfermedades Carenciales/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/efectos de los fármacos , Insulina/sangre , Selenio/deficiencia , Compuestos de Sodio/toxicidad , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Enfermedades Carenciales/sangre , Enfermedades Carenciales/etiología , Dieta , Modelos Animales de Enfermedad , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL
13.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34290138

RESUMEN

Nuclear envelope budding (NEB) is a recently discovered alternative pathway for nucleocytoplasmic communication distinct from the movement of material through the nuclear pore complex. Through quantitative electron microscopy and tomography, we demonstrate how NEB is evolutionarily conserved from early protists to human cells. In the yeast Saccharomyces cerevisiae, NEB events occur with higher frequency during heat shock, upon exposure to arsenite or hydrogen peroxide, and when the proteasome is inhibited. Yeast cells treated with azetidine-2-carboxylic acid, a proline analog that induces protein misfolding, display the most dramatic increase in NEB, suggesting a causal link to protein quality control. This link was further supported by both localization of ubiquitin and Hsp104 to protein aggregates and NEB events, and the evolution of these structures during heat shock. We hypothesize that NEB is part of normal cellular physiology in a vast range of species and that in S. cerevisiae NEB comprises a stress response aiding the transport of protein aggregates across the nuclear envelope.


Asunto(s)
Ácido Azetidinocarboxílico/toxicidad , Respuesta al Choque Térmico , Membrana Nuclear/fisiología , Pliegue de Proteína , Proteostasis/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/crecimiento & desarrollo , Arsenitos/toxicidad , Peróxido de Hidrógeno/toxicidad , Membrana Nuclear/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/metabolismo , Compuestos de Sodio/toxicidad , Ubiquitina/metabolismo , Ubiquitinación
14.
Toxicol Lett ; 350: 185-193, 2021 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-34303791

RESUMEN

A therapeutic strategy for prostate cancer (PCa) involves the use of 9-cis-retinoic acid (9cRA) to induce cancer stem cells (CSCs) differentiation and apoptosis. Polyinosinic:polycytidylic acid (PIC) is a Toll-like receptor 3 (TLR3) agonist that induces tumor cells apoptosis after activation. PIC+9cRA combination activates retinoic acid receptor ß (RARß) re-expression, leading to CSC differentiation and growth arrest. Since inorganic arsenic (iAs) targets prostatic stem cells (SCs), we hypothesized that arsenic-transformed SCs (As-CSCs) show an impaired TLR3-associated anti-tumor pathway and, therefore, are unresponsive to PIC activation. We evaluated TLR3-mediated activation of anti-tumor pathway based in RARß expression, on As-CSC and iAs-transformed epithelial cells (CAsE-PE). As-CSCs and CAsE-PE showed lower TLR3 and RARß basal expression compared to their respective isogenic controls WPE-Stem and RWPE-1. Also, iAs transformants showed reduced expression of mediators in TLR3 pathway. Importantly, As-CSCs were irresponsive to PIC+9cRA in terms of increased RARß and decreased SC-markers expression, while CAsE-PE, a heterogeneous cell line having a small SC population, were partially responsive. These observations indicate that iAs can impair TLR3 expression and anti-tumor pathway activated by PIC+9cRA in SCs and prostatic epithelial cells. These findings suggest that TLR3-activation based therapy may be an ineffective therapeutic alternative for iAs-associated PCa.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Compuestos de Sodio/toxicidad , Receptores Toll-Like/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Variación Genética , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/fisiopatología , Compuestos de Sodio/metabolismo , Receptores Toll-Like/metabolismo
16.
Toxicol Lett ; 349: 40-50, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34118311

RESUMEN

Previously, we reported that prolonged arsenic exposure impaired neuronal insulin signaling. Here we have further identified novel molecular mechanisms underlying neuronal insulin signaling impairment by arsenic. Arsenic treatment altered insulin dose-response curve and reduced maximum insulin response in differentiated human neuroblastoma SH-SY5Y cells, suggesting that arsenic hindered neuronal insulin signaling in a non-competitive like manner. Mechanistically, arsenic suppressed insulin receptor (IR) kinase activity, as witnessed by a decreased insulin-activated autophosphorylation of IR at Y1150/1151. Arsenic decreased the level of insulin receptor substrate 1 (IRS1) but increased the protein ratio between PI3K regulatory subunit, p85, and PI3K catalytic subunit, p110. Interestingly, co-immunoprecipitation demonstrated that arsenic did not alter a level of PI3K-p110/PI3K-p85 complex while increased PI3K-p85 levels in a PI3K-p110 depletion supernatant resulted from PI3K-p110 immunoprecipitation. These results indicated that arsenic increased PI3K-p85 which was free from PI3K-p110 binding. In addition, arsenic significantly increased interaction between IRS1 and PI3K-p85 but not PI3K-p110, suggesting that there may be a fraction of free PI3K-p85 interacting with IRS1. In vitro PI3K activity demonstrated that arsenic lowered PI3K activity in both basal and insulin-stimulated conditions. These results suggested that the increase in free PI3K-p85 by arsenic might compete with PI3K heterodimer for the same IRS1 binding site, in turn blocking the activation of its catalytic subunit, PI3K-p110. Taken together, our results provide additional insights into mechanisms underlying the impairment of neuronal insulin signaling by arsenic through the reduction of IR autophosphorylation, the increase in free PI3K-p85, and the impeding of PI3K activity.


Asunto(s)
Arsenitos/toxicidad , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Insulina/farmacología , Neuronas/efectos de los fármacos , Compuestos de Sodio/toxicidad , Antígenos CD/metabolismo , Sitios de Unión , Línea Celular Tumoral , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neuronas/enzimología , Neuronas/patología , Fosforilación , Unión Proteica , Receptor de Insulina/agonistas , Receptor de Insulina/metabolismo , Transducción de Señal
17.
Toxicol Appl Pharmacol ; 422: 115561, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33957193

RESUMEN

Arsenic is a global health concern that causes toxicity through ingestion of contaminated water and food. In vitro studies suggest that arsenic reduces stem and progenitor cell differentiation. Thus, this study determined if arsenic disrupted intestinal stem cell (ISC) differentiation, thereby altering the number, location, and/or function of intestinal epithelial cells. Adult male C57BL/6 mice were exposed to 0 or 100 ppb sodium arsenite (AsIII) through drinking water for 5 weeks. Duodenal sections were collected to assess changes in morphology, proliferation, and cell types. qPCR analysis revealed a 40% reduction in Lgr5 transcripts, an ISC marker, in the arsenic-exposed mice, although there were no changes in the protein expression of Olfm4. Secretory cell-specific transcript markers of Paneth (Defa1), Goblet (Tff3), and secretory transit amplifying (Math1) cells were reduced by 51%, 44%, and 30% respectively, in the arsenic-exposed mice, indicating significant impacts on the Wnt-dependent differentiation pathway. Further, protein levels of phosphorylated ß-catenin were reduced in the arsenic-exposed mice, which increased the expression of Wnt-dependent transcripts CD44 and c-myc. PCA analysis, followed by MANOVA and regression analyses, revealed significant changes and correlations between Lgr5 and the transit amplifying (TA) cell markers Math1 and Hes1, which are in the secretory cell pathway. Similar comparisons between Math1 and Defa1 show that terminal differentiation into Paneth cells is also reduced in the arsenic-exposed mice. The data suggests that ISCs are not lost following arsenic exposure, but rather, specific Wnt-dependent progenitor cell formation and terminal differentiation in the small intestine is reduced.


Asunto(s)
Arsenitos/toxicidad , Diferenciación Celular/efectos de los fármacos , Duodeno/efectos de los fármacos , Células de Paneth/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Compuestos de Sodio/toxicidad , Células Madre/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación hacia Abajo , Duodeno/metabolismo , Duodeno/patología , Masculino , Ratones Endogámicos C57BL , Células de Paneth/metabolismo , Células de Paneth/patología , Receptores Acoplados a Proteínas G/genética , Células Madre/metabolismo , Células Madre/patología , Factor Trefoil-3/genética , Factor Trefoil-3/metabolismo , Vía de Señalización Wnt , alfa-Defensinas/genética , alfa-Defensinas/metabolismo
18.
BMC Pharmacol Toxicol ; 22(1): 19, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33827703

RESUMEN

BACKGROUND: Arsenic poisoning affects millions of people. The inorganic forms of arsenic are more toxic. Treatment for arsenic poisoning relies on chelation of extracellularly circulating arsenic molecules by 2,3-dimecaptosuccinic acid (DMSA). As a pharmacological intervention, DMSA is unable to chelate arsenic molecules from intracellular spaces. The consequence is continued toxicity and cell damage in the presence of DMSA. A two-pronged approach that removes extracellular arsenic, while protecting from the intracellular arsenic would provide a better pharmacotherapeutic outcome. In this study, Coenzyme Q10 (CoQ10), which has been shown to protect from intracellular organic arsenic, was administered separately or with DMSA; following oral exposure to sodium meta-arsenite (NaAsO2) - a very toxic trivalent form of inorganic arsenic. The aim was to determine if CoQ10 alone or when co-administered with DMSA would nullify arsenite-induced toxicity in mice. METHODS: Group one represented the control; the second group was treated with NaAsO2 (15 mg/kg) daily for 30 days, the third, fourth and fifth groups of mice were given NaAsO2 and treated with 200 mg/kg CoQ10 (30 days) and 50 mg/kg DMSA (5 days) either alone or in combination. RESULTS: Administration of CoQ10 and DMSA resulted in protection from arsenic-induced suppression of RBCs, haematocrit and hemoglobin levels. CoQ10 and DMSA protected from arsenic-induced alteration of WBCs, basophils, neutrophils, monocytes, eosinophils and platelets. Arsenite-induced dyslipidemia was nullified by administration of CoQ10 alone or in combination with DMSA. Arsenite induced a drastic depletion of the liver and brain GSH; that was significantly blocked by CoQ10 and DMSA alone or in combination. Exposure to arsenite resulted in significant elevation of liver and kidney damage markers. The histological analysis of respective organs confirmed arsenic-induced organ damage, which was ameliorated by CoQ10 alone or when co-administered with DMSA. When administered alone, DMSA did not prevent arsenic-driven tissue damage. CONCLUSIONS: Findings from this study demonstrate that CoQ10 and DMSA separately or in a combination, significantly protect against arsenic-driven toxicity in mice. It is evident that with further pre-clinical and clinical studies, an adjunct therapy that incorporates CoQ10 alongside DMSA may find applications in nullifying arsenic-driven toxicity.


Asunto(s)
Antídotos/uso terapéutico , Intoxicación por Arsénico/tratamiento farmacológico , Arsenitos/toxicidad , Quelantes/uso terapéutico , Sustancias Protectoras/uso terapéutico , Compuestos de Sodio/toxicidad , Succímero/uso terapéutico , Ubiquinona/análogos & derivados , Animales , Intoxicación por Arsénico/sangre , Intoxicación por Arsénico/metabolismo , Intoxicación por Arsénico/patología , Células Sanguíneas/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Quimioterapia Combinada , Glutatión/metabolismo , Hematócrito , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ubiquinona/uso terapéutico
19.
Reprod Toxicol ; 102: 1-9, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33766721

RESUMEN

Arsenic (As) is an endocrine disrupting chemical that can disturb the male reproductive system. In a previous study, it was suggested that testicular macrophages could display a role in endocrine disruption induced by As exposure. This work aimed to evaluate the effects of chronic As exposure in the testis function of Wistar rats and examine the participation of macrophage activation and inflammatory response in these processes. We examined gene expression of steroidogenic machinery and immunological markers by RT-QPCR, plasma testosterone concentrations, sperm count and morphology, and histomorphometrical parameters after 60-days exposure to 1 or 5 mg.kg-1.day-1 of sodium arsenite, combined or not with 50 µg.kg-1 of LPS administered one day before euthanasia. We have demonstrated that As exposure reduced the weight of androgen-dependent organs and induced changes in spermatogenesis, in particular at the highest dose. LPS and As co-exposure promoted a decrease in testosterone synthesis, but did not increase the overexpression of markers of macrophage activation seen in LPS-only rats. Our results suggest that As does not alter the testicular macrophage function, but under immunological challenges LPS and As can display a complex interaction, which could lead to endocrine disruption.


Asunto(s)
Arsenitos/toxicidad , Disruptores Endocrinos/toxicidad , Compuestos de Sodio/toxicidad , Testículo/efectos de los fármacos , Animales , Arsénico/metabolismo , Disruptores Endocrinos/metabolismo , Activación de Macrófagos , Masculino , Ratas , Ratas Wistar , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Testículo/metabolismo , Testosterona/sangre
20.
Environ Health Prev Med ; 26(1): 34, 2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33706700

RESUMEN

BACKGROUND: Arsenic is a developmental neurotoxicant. It means that its neurotoxic effect could occur in offspring by maternal arsenic exposure. Our previous study showed that developmental arsenic exposure impaired social behavior and serotonergic system in C3H adult male mice. These effects might affect the next generation with no direct exposure to arsenic. This study aimed to detect the social behavior and related gene expression changes in F2 male mice born to gestationally arsenite-exposed F1 mice. METHODS: Pregnant C3H/HeN mice (F0) were given free access to tap water (control mice) or tap water containing 85 ppm sodium arsenite from days 8 to 18 of gestation. Arsenite was not given to F1 or F2 mice. The F2 mice were generated by mating among control F1 males and females, and arsenite-F1 males and females at the age of 10 weeks. At 41 weeks and 74 weeks of age respectively, F2 males were used for the assessment of social behavior by a three-chamber social behavior apparatus. Histological features of the prefrontal cortex were studied by ordinary light microscope. Social behavior-related gene expressions were determined in the prefrontal cortex by real time RT-PCR method. RESULTS: The arsenite-F2 male mice showed significantly poor sociability and social novelty preference in both 41-week-old group and 74-week-old group. There was no significant histological difference between the control mice and the arsenite-F2 mice. Regarding gene expression, serotonin receptor 5B (5-HT 5B) mRNA expression was significantly decreased (p < 0.05) in the arsenite-F2 male mice compared to the control F2 male mice in both groups. Brain-derived neurotrophic factor (BDNF) and dopamine receptor D1a (Drd1a) gene expressions were significantly decreased (p < 0.05) only in the arsenite-F2 male mice of the 74-week-old group. Heme oxygenase-1 (HO-1) gene expression was significantly increased (p < 0.001) in the arsenite-F2 male mice of both groups, but plasma 8-hydroxy-2'-deoxyguanosine (8-OHdG) and cyclooxygenase-2 (COX-2) gene expression were not significantly different. Interleukin-1ß (IL-1ß) mRNA expression was significantly increased only in 41-week-old arsenite-F2 mice. CONCLUSIONS: These findings suggest that maternal arsenic exposure affects social behavior in F2 male mice via serotonergic system in the prefrontal cortex. In this study, COX-2 were not increased although oxidative stress marker (HO-1) was increased significantly in arsnite-F2 male mice.


Asunto(s)
Arsénico/toxicidad , Arsenitos/toxicidad , Contaminantes Ambientales/toxicidad , Expresión Génica/efectos de los fármacos , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Conducta Social , Compuestos de Sodio/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Femenino , Marcadores Genéticos , Masculino , Ratones , Ratones Endogámicos C3H , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Corteza Prefrontal/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/psicología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serotonina/genética , Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA