Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Environ Pollut ; 346: 123651, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38408505

RESUMEN

Triphenyl phosphate (TPHP) is an organophosphorus flame retardant, but its cardiac toxicity has not been adequately investigated. Therefore, in the current study, the effect of TPHP on the heart and the underlying mechanism involved was evaluated. C57BL/6 J mice were administered TPHP (0, 5, and 50 mg/kg/day) for 30 days. In addition, H9c2 cells were treated with three various concentrations (0, 50, and 150 µM) of TPHP, with and without the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine or the mitochondrial fusion promoter M1. TPHP caused cardiac fibrosis and increased the levels of CK-MB and LDH in the serum. TPHP increased the levels of ROS, malondialdehyde (MDA), and decreased the level of superoxide dismutase (SOD) and Glutathione peroxidase (GSH-Px). Furthermore, TPHP caused mitochondrial damage, and induced fusion and fission disorders that contributed to mitophagy in both the heart of C57BL/6 J mice and H9c2 cells. Transcriptome analysis showed that TPHP induced up- or down-regulated expression of various genes in myocardial tissue and revealed enriched apoptosis pathways. It was also found that TPHP could remarkably increase the expression levels of Bax, cleaved Caspase-9, cleaved Caspase-3, and decreased Bcl-2, thereby causing apoptosis in H9c2 cells. Taken together, the results suggested that TPHP promoted mitophagy through mitochondria fusion dysfunction resulting from oxidative stress, leading to fibrosis by inducing myocardial apoptosis.


Asunto(s)
Retardadores de Llama , Miocitos Cardíacos , Organofosfatos , Ratones , Animales , Cardiotoxicidad/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retardadores de Llama/metabolismo , Mitofagia , Ratones Endogámicos C57BL , Compuestos Organofosforados/metabolismo , Estrés Oxidativo , Apoptosis , Fibrosis
2.
Chem Biol Interact ; 390: 110870, 2024 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-38220133

RESUMEN

Busulfan, a bifunctional alkylated chemotherapeutic agent, has male reproductive toxicity and induce oligospermia, which is associated with ferroptosis. However, the specific target cells of busulfan-induced oligospermia triggered by ferroptosis are largely elusive, and the detailed mechanisms also require further exploration. In the present study, busulfan (0.6, and 1.2 mM, 48 h) causes ferroptosis in GC-1 spg cells through inducing Fe2+, ROS and MDA accumulation and functional inhibition of Xc-GSH-GPX4 antioxidant system. After inhibition of ferroptosis by Fer-1 (1 µM, pretreatment for 2 h) or DFO (10 µM, pretreatment for 2 h) reverses busulfan-induced destructive effects in GC-1 spg cells. Furthermore, using RNA-seq and Western blotting, we found that busulfan promotes autophagy-dependent ferritin degradation, as reflected by enriching in autophagy, increased LC3 II, Beclin1 and NCOA4, as well as decreased P62 and ferritin heavy chain 1 (FTH1). Ultimately, GC-1 spg cells and Balb/c mice were treated with busulfan and/or 3-MA, the inhibitor of autophagy. The results displayed that inhibition of autophagy relieves busulfan-induced FTH1 degradation and then blocks the occurrence of ferroptosis in GC-1 spg cells and testicular spermatogonia, which subsequently alleviates busulfan-caused testicular damage and spermatogenesis disorders. In summary, these data collectively indicated that ferroptosis of spermatogonia is involved in busulfan-induced oligospermia and mediated by autophagy-dependent FTH1 degradation, identifying a new target for the therapy of busulfan-induced male infertility.


Asunto(s)
Acetatos , Ferroptosis , Oligospermia , Fenoles , Humanos , Masculino , Animales , Ratones , Busulfano/toxicidad , Espermatogonias , Oligospermia/inducido químicamente , Autofagia
4.
Ecotoxicol Environ Saf ; 264: 115459, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37703808

RESUMEN

Aluminum is a neurotoxic food contaminant. Aluminum trichloride (AlCl3) causes hippocampal mitochondrial damage, leading to hippocampal injury. Damaged mitochondria can release mitochondrial reactive oxygen species (mtROS) and activate nucleotide-binding oligomerization domain-like receptor-containing 3 (NLRP3) inflammasomes and apoptosis. E3 ubiquitin ligase PARK2 (Parkin)-mediated mitophagy can attenuate mitochondrial damage. However, the role of mitophagy in AlCl3-induced mice hippocampal damage and its regulatory mechanism remain elusive. First, C57BL/6 N mice were treated with 0, 44.825, 89.65, and 179.3 mg/kg body weight AlCl3 drinking water for 90 d. Apoptosis, NLRP3-inflammasome activation and mitochondrial damage were increased in AlCl3-induced hippocampal damage. In addition, Parkin-mediated mitophagy peaked in the middle-dose group and was slightly attenuated in the high-dose group. Subsequently, we used wild-type and Parkin knockout (Parkin-/-) mice to investigate the AlCl3-induced hippocampal damage. The results showed that Parkin-/- inhibited mitophagy, and aggravated AlCl3-induced mitochondrial damage, NLRP3-inflammasome activation, apoptosis and hippocampal damage. Finally, we administered MitoQ (mtROS inhibitor) and MCC950 (NLRP3 inhibitor) to AlCl3-treated Parkin-/- mice to investigate the mechanism of Parkin-mediated mitophagy. The results showed that inhibition of mtROS and NLRP3 attenuated hippocampal NLRP3-inflammasome activation, apoptosis, and damage in AlCl3-treated Parkin-/- mice. These findings indicate that Parkin-mediated mitophagy protects against AlCl3-induced hippocampal apoptosis in mice via the mtROS-NLRP3 pathway.


Asunto(s)
Cloruro de Aluminio , Hipocampo , Inflamasomas , Mitofagia , Animales , Ratones , Cloruro de Aluminio/toxicidad , Apoptosis , Hipocampo/efectos de los fármacos , Hipocampo/patología , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética
5.
Ecotoxicol Environ Saf ; 256: 114876, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37027944

RESUMEN

Triphenyl phosphate (TPHP) is a widely used organophosphate flame retardant and has biological toxicity. Previous studies showed TPHP can restrain testosterone biosynthesis in Leydig cells, while the underlying mechanisms remain unclear. In this study, C57BL/6J male mice were exposed to 0, 5, 50, and 200 mg/kg B.W. of TPHP for 30 d by oral, as well as TM3 cells were treated with 0, 50, 100, and 200 µM of TPHP for 24 h. Results showed that TPHP induced testes damage, including spermatogenesis disorders and testosterone synthesis inhibition. Meanwhile, TPHP can cause apoptosis in testicular Leydig cells and TM3 cells, as evidenced by the increased apoptosis rate and decreased Bcl-2/Bax ratio. Moreover, TPHP disrupted mitochondrial ultrastructure of testicular Leydig cells and TM3 cells, reduced healthy mitochondria content and depressed mitochondrial membrane potential of TM3 cells, as well as inhibited mitochondrial fusion proteins mitofusin 1 (Mfn1), mitofusin 2 (Mfn2), and optic atrophy 1 (Opa1) expression, without effect on mitochondrial fission proteins dynamin-related protein 1 (Drp1) and fission 1 (Fis1) in testicular tissue and/or TM3 cells. Then, the mitochondrial fusion promoter M1 was used to pre-treat TPHP-exposed TM3 cells to determine the roles of mitochondrial fusion inhibition in TPHP-induced Leydig cells apoptosis. The results showed M1 pretreatment alleviated the above changes and further mitigated TM3 cells apoptosis and testosterone levels decreased, indicating TPHP induced TM3 cells apoptosis by inhibited mitochondrial fusion. Intriguingly, the intervention experiment of N-acetylcysteine (NAC) showed that TPHP-induced mitochondrial fusion inhibition is ROS dependent, because inhibition of ROS overproduction alleviated mitochondrial fusion inhibition, and subsequently relieved TPHP-induced apoptosis in TM3 cells. In summary, above data revealed that apoptosis is a specific mechanism for TPHP-induced male reproductive toxicity, and that ROS-mediated mitochondrial fusion inhibition is responsible for Leydig cells apoptosis caused by TPHP.


Asunto(s)
Células Intersticiales del Testículo , Dinámicas Mitocondriales , Ratones , Animales , Masculino , Células Intersticiales del Testículo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos C57BL , Apoptosis , Proteínas Mitocondriales/metabolismo , Organofosfatos/metabolismo , Testosterona/metabolismo
6.
Heliyon ; 8(11): e11767, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36451752

RESUMEN

Aims: Inflammatory cytokines can destroy the immune balance and lead to adverse pregnancy outcomes. Benzo (a) pyrene (BaP) may induce premature delivery through leading inflammatory reaction. We screened out inflammatory factors related to adverse pregnancy outcomes through bioinformatics analysis. Then we verified the correlation between adverse pregnancy outcomes caused by BaP and abnormal expression of those inflammatory factors. Main methods: The Gene Expression Omnibus (GEO) database was used to analyze by R to screen the inflammatory genes related to adverse pregnancy outcomes. Based on the established BaP exposure animal model, the expression of key cytokines in placenta was detected by immunohistochemistry. Key findings: According to the data analysis of GEO database, the expression of IL18, IL18BP and IL18R was up-regulated, while the expression of IL1RN was down regulated in the adverse pregnancy outcome group. BaP exposure significantly increased the rate of adverse pregnancy outcome in pregnant golden hamsters, and also significantly interferes with the process of embryonic development. Meanwhile, the expression of IL18, IL18BP and IL18R in placenta was increased, while the expression of IL1RN protein was decreased, consistent with the mRNA expression level gathered by bioinformatics analysis. Significance: BaP may induce the inflammatory reaction to cause adverse pregnancy outcome by regulating the expression of IL18, IL18BP, IL18R and IL1RN. Our findings provide experimental basis for the prevention of adverse pregnancy outcome caused by BaP.

8.
Toxicology ; 473: 153193, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35533795

RESUMEN

Busulfan, a chemotherapeutic agent for cancer, has detrimental effects on germ cells and fertility, yet the specific mechanisms remain largely uncertain. The blood-testis barrier (BTB) maintains a suitable microenvironment for germ cells self-renewal and spermatogenesis by blocking the interference and damage of deleterious substances. Therefore, we hypothesized that BTB abnormalities might be involved in busulfan-induced oligospermia. To verify the hypothesis, thirty male Balb/c mice were randomly administered with busulfan (at a total dose of 40 mg/kg body weight) by intraperitoneal injection for 4 weeks to establish the model of oligospermia. The results displayed that busulfan caused testicular histopathological lesions and spermatogenesis disorder. Meanwhile, busulfan disrupted BTB integrity and lessened the expressions of BTB junction proteins, including Occludin, Claudin-11 and Connexin-43. Furthermore, busulfan activated the endoplasmic reticulum (ER) stress and PERK-eIF2α signaling pathway, reflected by the increased protein expressions of GRP78, p-PERK, p-eIF2α, ATF4 and CHOP. Finally, to evaluate whether the ER stress is involved in busulfan-induced BTB destruction, the ER stress inhibitor 4-Phenylbutyric acid (4-PBA, 1 mM) was used to intervene in busulfan-exposed TM4 cells. The results displayed that inhibition of ER stress alleviated the reduction of BTB junction protein expressions induced by busulfan in TM4 cells. These data collectively indicated that busulfan-induced BTB impairment was mediated by triggering ER stress and activation of the PERK-eIF2α signaling pathway, thereby damaging the spermatogenesis, providing a new therapeutic target for male infertility induced by busulfan.


Asunto(s)
Factor 2 Eucariótico de Iniciación , Oligospermia , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis , Barrera Hematotesticular/metabolismo , Busulfano/toxicidad , Estrés del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Masculino , Ratones , Transducción de Señal , eIF-2 Quinasa/metabolismo
9.
Environ Toxicol Pharmacol ; 90: 103794, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34971797

RESUMEN

Many studies have shown that aflatoxin B1 (AFB1) can cause cytotoxicity in numerous cells and organs induced by oxidative stress. However, the toxic effects and related mechanism of AFB1 on the microglia cells in the spinal cords have not been studied yet. Our results showed that AFB1 significantly reduced the number of microglia cells, increased the oxidants (malonaldehyde and hydrogen peroxide) but decreased the anti-oxidants (superoxide dismutase and total antioxidant capacity) in a dose dependent manner in mice spinal cords. In addition, AFB1 significantly increased the oxidative stress, promoted apoptosis and cell cycle arrest in G2-M phase, and activated NF-κB phosphorylation in BV2 microglia cells. However, the addition of active oxygen scavenger N-acetylcysteine can significantly reduce the ROS production, improve cell cycle arrest, reduce apoptosis, and the expression of phosphorylated NF-κB in BV2 microglia cells. These results indicate that AFB1 induces microglia cells apoptosis through oxidative stress by activating NF-κB signaling pathway.


Asunto(s)
Aflatoxina B1/toxicidad , Apoptosis/efectos de los fármacos , Microglía/efectos de los fármacos , Acetilcisteína/administración & dosificación , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Células Cultivadas , Masculino , Ratones , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Médula Espinal/efectos de los fármacos
10.
Psychopharmacology (Berl) ; 238(10): 2923-2936, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34231002

RESUMEN

Post-traumatic stress disorder (PTSD) is a serious stress-related neuropsychiatric disorder caused by major traumatic events. Abnormal activity of the locus coeruleus (LC)-noradrenergic system is related to the development of PTSD-like symptoms. Our previous studies have indicated that endoplasmic reticulum (ER) stress induced neuronal apoptosis of LC in rats with PTSD. The purpose of this study was to further investigate the role of ER stress pathways in LC neuronal dysfunction and elucidate the effect of the bioactive component tetramethylpyrazine (TMP) against ER stress response. We used an acute exposure to single prolonged stress (SPS) to model PTSD in rats. There were higher norepinephrine (NE) levels in the brain, increased tyrosine hydroxylase expression in LC, and enhanced anxiety-like behaviors in rats exposed to SPS, which were observed by enzyme-linked immunosorbent assay, western blot analysis and elevated plus maze test, respectively. In addition, the three major pathways of ER stress were activated by SPS exposure, which may be involved in the dysregulation of the LC-noradrenergic system of rats with PTSD. Furthermore, we found that TMP administration significantly suppressed the increased responsiveness of LC-noradrenergic system, effectively reduced the anxiety response of SPS rats, and selectively attenuated the activation of pro-apoptotic ER stress pathways. The results suggest that TMP was efficient in improving the LC-NE dysfunction induced by excessive ER stress. TMP exhibited a significant neuroprotective effect and potential therapeutics on PTSD-like symptoms.


Asunto(s)
Locus Coeruleus , Trastornos por Estrés Postraumático , Animales , Apoptosis , Pirazinas/farmacología , Ratas , Trastornos por Estrés Postraumático/tratamiento farmacológico
11.
Ecotoxicol Environ Saf ; 216: 112213, 2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33838459

RESUMEN

Aflatoxin B1 (AFB1) pollutes foodstuffs and feeds, causing a food safety problem and seriously endangering human and animal health. Liver is the principal organ for AFB1 accumulation and biotransformation, during which AFB1 can cause acute and chronic liver damage, however, the specific mechanism is not completely clear. Mitochondria are the primary organelle of cellular bio-oxidation, providing 95% energy for liver to execute its multiple functions. Therefore, we speculated that mitochondrial dysfunction is involved in AFB1-induced liver injury. To verify the hypothesis, a total of eighty healthy male mice were randomly divided into four groups on average, and exposed with 0, 0.375, 0.75 and 1.5 mg/kg body weight AFB1 by intragastric administration for 30 d. The results displayed that AFB1 triggered liver injury accompanied by oxidative stress. AFB1 exposure also damaged mitochondria structure, decreased mitochondrial membrane potential (MMP), as well as increased cytoplasmic cytochrome c (Cyt-c) protein expression, Bax, p53, Caspase-3/9 protein and/or mRNA expression levels and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine-5'-triphosphate (dUTP) nick end labeling (TUNEL) staining positive cells in mice liver. Meanwhile, AFB1 exposure elevated pyruvate content, inhibited tricarboxylic acid (TCA) cycle rate-limiting enzymes and electron transport chain (ETC) complexes I-V activities, disturbed ETC complexes I-V subunits mRNA expression levels and reduced adenosine triphosphate (ATP) level in mice liver. These results indicated that AFB1 destroyed mitochondrial structure, activated mitochondrion-dependent apoptosis and induced mitochondrial dysfunction. In addition, AFB1 disrupted mitochondrial biogenesis, presented as the abnormalities of protein and/or gene expression levels of voltage dependent anion channel protein 1 (VDAC1), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), nuclear respiratory factor 1 (Nrf1) and mitochondrial transcription factor A (Tfam). This may contribute to hepatic and mitochondrial lesions induced by AFB1. These results provide a new perspective for elucidating the mechanisms of AFB1 hepatotoxicity.

12.
Environ Sci Pollut Res Int ; 28(35): 47921-47931, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33895957

RESUMEN

As a persistent pollutant, microplastics (MPs) have been reported to induce sperm quantity decrease in mice. However, the related mechanism remains obscure. Therefore, this study is intended to explore the effects of polystyrene microplastics (PS-MPs) on male reproduction and its related mechanism of blood-testis barrier (BTB) impairment. Thirty-two adult male Wistar rats were divided randomly into four groups fed with PS-MPs for 90 days at doses of 0 mg/day (control group), 0.015 mg/day, 0.15 mg/day, and 1.5 mg/day, respectively. The present results have shown that PS-MP exposure led to the damage of seminiferous tubule, resulted in apoptosis of spermatogenic cells, and decreased the motility and concentration of sperm, while the abnormality of sperm was elevated. Meanwhile, PS-MPs could induce oxidative stress and activate the p38 MAPK pathway and thus deplete the nuclear factor erythroid-2 related factor 2 (Nrf2). Noteworthily, PS-MPs led to the BTB-related protein expression decrease. All these results demonstrated that PS-MP exposure may lead to the destruction of BTB integrity and the apoptosis of spermatogenic cells through the activation of the MAPK-Nrf2 pathway. The current study provided novelty evidence for elucidating the effects of PS-MPs on male reproductive toxicity and its potential mechanism.


Asunto(s)
Microplásticos , Poliestirenos , Animales , Barrera Hematotesticular , Masculino , Ratones , Factor 2 Relacionado con NF-E2 , Plásticos , Ratas , Ratas Wistar , Transducción de Señal
13.
Toxicology ; 449: 152665, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359712

RESUMEN

Microplastics (MPs) are receiving increased attention as a harmful environmental pollutant. Studies have investigated that MPs have reproductive toxicity, but the mechanism is little known. Here, we aimed to investigate the effects of polystyrene microplastics (PS-MPs) on ovary in rats and the underlying molecular mechanisms. in vivo, thirty-two female Wistar rats were exposed to 0.5 µm PS-MPs at different concentrations (0, 0.015, 0.15 and 1.5 mg/d) for 90 days. And then, all animals were sacrificed, ovaries and blood were collected for testing. in vitro, granulosa cells (GCs) were separated from rat ovary and treated with 0、1、5、25 µg/mL PS-MPs and reactive oxygen species (ROS) inhibitor N-Acetyl-l-cysteine (NAC) respectively. Our results showed that PS-MPs could enter into GCs and result in the reducing of growing follicles number. And the Enzyme-linked immunosorbent assay (ELISA) manifested that PS-MPs could obviously decrease the level of anti-Müllerian hormone (AMH). In addition, PS-MPs induced oxidative stress, apoptosis of GCs and ovary fibrosis evidenced by assay kits, flow cytometry, immunohistochemistry, Masson's trichrome and Sirius red staining. Moreover, the western blot assay manifested that PS-MPs exposure significantly increased the expression levels of Wnt/ß-Catenin signaling pathways-related proteins (Wnt, ß-catenin, p-ß-catenin) and the main fibrosis markers (transforming growth factor-ß (TGF-ß), fibronectin, α-smooth muscle actin (α-SMA). Additionally, the expression levels of Wnt and p-ß-catenin, apoptosis of GCs decreased after NAC treatment. In summary, polystyrene microplastics cause fibrosis via Wnt/ß-Catenin signaling pathway activation and granulosa cells apoptosis of ovary through oxidative stress in rats, both of which ultimately resulted in decrease of ovarian reserve capacity.


Asunto(s)
Apoptosis/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Microplásticos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Poliestirenos/toxicidad , Animales , Apoptosis/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Fibrosis/patología , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Ovario/efectos de los fármacos , Ovario/patología , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar
14.
Res Vet Sci ; 129: 103-108, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31954314

RESUMEN

To evaluate the protective role of lycopene (LYC) against aflatoxin B1 (AFB1)-induced erythrocyte dysfunction and oxidative stress, male kunming mice were treated with LYC (5 mg/kg) and/or AFB1 (0.75 mg/kg) by intragastric administration for 30 d. Hematological indices were detected to assess erythrocyte function. The erythrocytes C3b receptor rate (E-C3bRR) and erythrocytes C3b immune complex rosette rate (E-ICRR) were detected to assess erythrocyte immune function. Hydrogen peroxide (H2O2) and malondialdehyde (MDA) contents and superoxide dismutase (SOD) and catalase (CAT) activities were determined to evaluate erythrocyte oxidative stress. The results showed that LYC administration significantly relieved AFB1-induced erythrocyte dysfunction by increasing the levels of red blood cell count (RBC), hemoglobin (HGB) and hematocrit (HCT), as well as reducing red blood cell volume distribution width (RDW) level, while the levels of mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC) and mean platelet volume (MPV) had no significant differences among the four groups. Besides, LYC ameliorated AFB1-induced erythrocyte immune dysfunction by increasing E-C3bRR and decreasing E-ICRR. Furthermore, LYC also alleviated AFB1-induced erythrocyte oxidative stress by decreasing H2O2 and MDA contents and increasing SOD and CAT activities. These results indicated that LYC protected against AFB1-induced erythrocyte dysfunction and oxidative stress in mice. The findings could lead a possible therapeutics for the management of AFB1-induced erythrocyte toxicity.


Asunto(s)
Aflatoxina B1/toxicidad , Antioxidantes/farmacología , Eritrocitos/efectos de los fármacos , Licopeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Índices de Eritrocitos , Hematócrito , Peróxido de Hidrógeno , Masculino , Malondialdehído , Ratones
15.
Biol Trace Elem Res ; 197(2): 580-590, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31848921

RESUMEN

Aluminum (Al) alters iron regulatory factors content and leads to the changes in iron-related proteins causing iron accumulation. But limited evidence ascertains this hypothesis. Therefore, our experiment was conducted and four groups of male Wistar rats were orally administrated of 0, 50, 150, and 450 mg/kg BW/d aluminum chloride (AlCl3) for 90 days by drinking water, respectively. The cognitive function, pathological lesion of hippocampus, oxidative stress, as well as iron-related proteins and iron regulatory factors expression were detected. The results showed that AlCl3 remarkably induced the oxidative stress and pathological lesion in the hippocampus and impaired the learning-memory ability. The contents of Al and iron increased in all AlCl3-exposed groups. Meanwhile, the increased divalent metal transporter 1 (DMT1) expression enhanced iron import and the decreased ferroportin 1 (Fpn1) expression reduced iron export in AlCl3-exposed groups. The iron accumulated and ferritin heavy chains (Fth) expression decreased in all AlCl3-exposed groups led to an increase in free iron. The study also showed that iron regulatory factor iron regulatory protein 2 (IRP2) was decreased and hepcidin was increased in AlCl3-exposed groups. The results indicated that AlCl3 induces iron dyshomeostasis presenting as iron accumulation, the disordered expression of iron import, export, store, and regulatory proteins in rat hippocampus accompanied with oxidative stress, pathological lesion, and impaired learning-memory ability.


Asunto(s)
Hipocampo , Hierro , Aluminio/toxicidad , Cloruro de Aluminio , Compuestos de Aluminio/toxicidad , Animales , Masculino , Estrés Oxidativo , Ratas , Ratas Wistar
16.
Food Funct ; 10(7): 3868-3879, 2019 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-31184641

RESUMEN

Lycopene (LYC) has been reported to exhibit antioxidant and immunoprotective activities, and our previous studies confirmed that LYC can alleviate multiple tissue damage induced by aflatoxin B1 (AFB1). However, it is unclear whether LYC could relieve the AFB1-induced immunosuppression. Thus, forty-eight male mice were randomly allocated and treated with LYC (5 mg kg-1) and/or AFB1 (0.75 mg kg-1) by intragastric administration for 30 days. We found that LYC alleviated AFB1-induced immunosuppression by relieving splenic structure injury and increasing the spleen weight, spleen coefficient, T lymphocyte subsets, the contents of IL-2, IFN-γ and TNF-α in serum, as well as the mRNA expression of IL-2, IFN-γ and TNF-α in spleen. Furthermore, LYC inhibited oxidative stress induced by AFB1via decreasing the levels of reactive oxygen species (ROS), hydrogen peroxide (H2O2) and malondialdehyde (MDA), while enhancing the total antioxidant capacity (T-AOC) and antioxidant enzyme activities. In addition, LYC also restrained splenic apoptosis through blocking mitochondria-mediated apoptosis in AFB1 intoxicated mice, presenting as the increase of mitochondrial membrane potential, and the decrease of cytoplasmic Cyt-c protein expression, cleaved Caspase-3 protein expression, Caspase-3/9 activities and mRNA expressions, as well as balancing the mitochondrial protein and mRNA expressions of Bax and Bcl-2. These results indicate that LYC can alleviate AFB1-induced immunosuppression by inhibiting oxidative stress and mitochondria-mediated apoptosis of mice spleen.


Asunto(s)
Aflatoxina B1/efectos adversos , Apoptosis/efectos de los fármacos , Terapia de Inmunosupresión , Licopeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Peróxido de Hidrógeno , Interferón gamma/sangre , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-2/sangre , Interleucina-2/genética , Interleucina-2/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bazo/lesiones , Bazo/patología , Linfocitos T , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
17.
Chem Biol Interact ; 299: 15-26, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30481499

RESUMEN

Many reports demonstrated that aluminum maltolate (Almal) has potential toxicity to human and animal. Our study has demonstrated that Almal can induce oxidative damage and apoptosis in PC12 cells and SH-SY5Y Cells, two in vitro models of neuronal cells. Hyperforin (HF) is a well-known antioxidant, anti-inflammatory, anti-amyloid and anti-depressant compound extracted from Hypericum perforatum extract. Here, we investigated the neuroprotective effect of HF against Almal-induced neurotoxicity in cultured PC12 cells and SH-SY5Y cells, mainly caused by oxidative stress. In the present study, HF significantly inhibited the formation of reactive oxygen species (ROS), decreased the level of lipid peroxide and enhanced the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) compared with Almal group in PC12 cells and SH-SY5Y cells. Additionally, HF suppressed the reduction of the mitochondrial membrane potential (MMP), cytochrome c (Cyt-c) release, activation of caspase-3, and the down-regulation of Bcl-2 expression and up-regulation of Bax expression induced by Almal in PC12 cells and SH-SY5Y cells. In summary, HF protects PC12 cells and SH-SY5Y cells from damage induced by Almal through reducing oxidative stress and preventing of mitochondrial-mediated apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Floroglucinol/análogos & derivados , Terpenos/farmacología , Animales , Caspasa 3/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Malondialdehído/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Compuestos Organometálicos/toxicidad , Células PC12 , Floroglucinol/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pironas/toxicidad , Ratas , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
18.
Chemosphere ; 209: 972-980, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30114748

RESUMEN

Aluminum (Al), a common environmental pollutant, has been reported to inhibit the immune functions of macrophage. However, the mechanisms involved remain unclear. In this study, murine peritoneal macrophages were exposed to 0, 0.27, 0.54, and 1.08 mg/mL of aluminium chloride (AlCl3) for 24 h, and then treated with 1 µg/mL lipopolysaccharide (LPS) for another 6 h. No addition of both AlCl3 and LPS serviced as control group. We observed that AlCl3 has cytotoxicity in murine peritoneal macrophages, showing a decrease in cell viability and an increase in lactate dehydrogenase release. Besides, AlCl3 exposure restrained the LPS-induced NLR pyrin domain containing 3 (NLRP3) inflammasome activation presented as NLRP3 expressions reduction, caspase-1 cleavage inhibition and interleukin 1 beta (IL-1ß) maturation lessened. Meanwhile, AlCl3 exposure decreased LPS-induced IKKß activity, IκBα phosphorylation, the phosphorylation and mRNA expression of NF-κB p65, as well the genes expression and concentration in medium supernatant of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). The results suggested that AlCl3 inhibited the activation of NF-κB signaling pathway induced by LPS, which maybe one of the upstream signals involved in the inhibition of NLRP3 inflammasome activation by AlCl3. This research can provide theoretical basis for understanding the immune toxicity of Al, and deepening the cognition of Al exposure hazards to immune response.


Asunto(s)
Inflamasomas/metabolismo , Interleucina-1beta/genética , Lipopolisacáridos/metabolismo , Macrófagos Peritoneales/metabolismo , Inhibidor NF-kappaB alfa/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Ratones , Inhibidor NF-kappaB alfa/metabolismo , Transducción de Señal
19.
Chemosphere ; 203: 170-178, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29614410

RESUMEN

Aluminum (Al) is a recognized environmental pollutant that causes neuroinflammatory lesions, leading to neurodegenerative diseases. Interleukin-1 (IL-1) signaling pathway is responsible for regulating inflammatory lesions. However, it remains unclear whether IL-1 signaling pathway is involved in neuroinflammatory lesions induced by Al exposure. In the present study, one hundred and twenty Wistar rats were orally exposed to 0, 50, 150 and 450 mg/kg BW/d aluminum trichloride (AlCl3) for 90 days, respectively. We found that AlCl3 exposure increased hippocampal Al concentration, reduced hippocampus coefficient, impaired cognitive ability, deteriorated microstructure of hippocampal CA1 and CA3 regions, increased reactive oxygen species (ROS) level, activated astrocytes and microglia, increased pro-inflammatory cytokines contents and mRNA expressions, and decreased anti-inflammatory cytokines contents and mRNA expressions in the hippocampus. These results indicated that AlCl3 induced the hippocampal inflammatory lesion (HIL). Moreover, AlCl3 exposure increased the mRNA and protein expression of IL-1 signaling pathway core components in the hippocampus, demonstrating that AlCl3 activated IL-1 signaling pathway. Furthermore, the correlation between interleukin-1ß (IL-1ß) content and HIL and activation of the IL-1 signaling pathway was analyzed. Results showed that IL-1ß content was positively correlated with pro-inflammatory cytokines contents and mRNA expressions and activation of IL-1 signaling pathway, and was negatively correlated with hippocampus coefficient, anti-inflammatory cytokines contents and mRNA expressions, and the number of hippocampal neurons. The above results demonstrate that AlCl3-induced HIL is associated with IL-1 signaling pathway, in which IL-1ß is a link.


Asunto(s)
Compuestos de Aluminio/toxicidad , Cloruros/toxicidad , Hipocampo/patología , Interleucina-1beta/metabolismo , Transducción de Señal , Aluminio/metabolismo , Cloruro de Aluminio , Animales , Citocinas/metabolismo , Hipocampo/metabolismo , Inflamación/inducido químicamente , Neuronas/metabolismo , Ratas , Ratas Wistar
20.
Biol Trace Elem Res ; 184(1): 173-185, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29027106

RESUMEN

Aluminum (Al) is known to induce apoptosis of osteoblasts (OBs). However, the mechanism is not yet established. To investigate the apoptotic mechanism of OBs induced by aluminum trichloride (AlCl3), the primary OBs from the craniums of fetal Wistar rats were exposed to 0 mg/mL (control group, CG), 0.06 mg/mL (low-dose group, LG), 0.12 mg/mL (mid-dose group, MG), and 0.24 mg/mL (high-dose group, HG) AlCl3 for 24 h, respectively. We observed that AlCl3 induced OB apoptosis with the appearance of apoptotic morphology and increase of apoptosis rate. Additionally, AlCl3 treatment activated mitochondrial-mediated signaling pathway, accompanied by mitochondrial membrane potential (ΔΨm) depolarization, release of cytochrome c from the mitochondria to the cytoplasm, as well as survival signal-related factor caspase-9 and caspase-3 activation. AlCl3 exposure also activated Fas/Fas ligand signaling pathway, presented as Fas, Fas ligand, and Fas-associated death domain expression enhancement and caspase-8 activation, as well as the hydrolysis of Bid to truncated Bid, suggesting that the Fas-mediated signaling pathway might aggravate mitochondria-mediated OB apoptosis through hydrolyzing Bid. Furthermore, AlCl3 exposure inhibited Bcl-2 protein expression and increased the expressions of Bax, Bak, and Bim in varying degrees. These results indicated that AlCl3 exposure induced OB apoptosis through activating Fas- and mitochondria-mediated signaling pathway and disrupted B-cell lymphoma-2 family proteins.


Asunto(s)
Compuestos de Aluminio/farmacología , Supervivencia Celular/efectos de los fármacos , Cloruros/farmacología , ARN Mensajero/metabolismo , Receptor fas/metabolismo , Cloruro de Aluminio , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Proteína Ligando Fas/metabolismo , Citometría de Flujo , Mitocondrias/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...