Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Environ Pollut ; 346: 123651, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38408505

RESUMO

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.


Assuntos
Retardadores de Chama , Miócitos Cardíacos , Organofosfatos , Camundongos , Animais , Cardiotoxicidade/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Retardadores de Chama/metabolismo , Mitofagia , Camundongos Endogâmicos C57BL , Compostos Organofosforados/metabolismo , Estresse Oxidativo , Apoptose , Fibrose
3.
Ecotoxicol Environ Saf ; 264: 115459, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37703808

RESUMO

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.


Assuntos
Cloreto de Alumínio , Hipocampo , Inflamassomos , Mitofagia , Animais , Camundongos , Cloreto de Alumínio/toxicidade , Apoptose , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
4.
Toxicology ; 473: 153193, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35533795

RESUMO

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.


Assuntos
Fator de Iniciação 2 em Eucariotos , Oligospermia , Fator 4 Ativador da Transcrição/metabolismo , Animais , Apoptose , Barreira Hematotesticular/metabolismo , Bussulfano/toxicidade , Estresse do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Masculino , Camundongos , Transdução de Sinais , eIF-2 Quinase/metabolismo
5.
Environ Toxicol Pharmacol ; 90: 103794, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34971797

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Apoptose/efeitos dos fármacos , Microglia/efeitos dos fármacos , Acetilcisteína/administração & dosagem , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Camundongos , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Medula Espinal/efeitos dos fármacos
6.
Ecotoxicol Environ Saf ; 216: 112213, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33838459

RESUMO

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.

7.
Toxicology ; 449: 152665, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33359712

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Microplásticos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Poliestirenos/toxicidade , Animais , Apoptose/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Fibrose/induzido quimicamente , Fibrose/metabolismo , Fibrose/patologia , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Ovário/efeitos dos fármacos , Ovário/patologia , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar
8.
Biol Trace Elem Res ; 197(2): 580-590, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31848921

RESUMO

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.


Assuntos
Hipocampo , Ferro , Alumínio/toxicidade , Cloreto de Alumínio , Compostos de Alumínio/toxicidade , Animais , Masculino , Estresse Oxidativo , Ratos , Ratos Wistar
9.
Food Funct ; 10(7): 3868-3879, 2019 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-31184641

RESUMO

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.


Assuntos
Aflatoxina B1/efeitos adversos , Apoptose/efeitos dos fármacos , Terapia de Imunossupressão , Licopeno/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio , Interferon gama/sangue , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-2/sangue , Interleucina-2/genética , Interleucina-2/metabolismo , Masculino , Malondialdeído/metabolismo , Camundongos , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Baço/lesões , Baço/patologia , Linfócitos T , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
10.
Chem Biol Interact ; 299: 15-26, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30481499

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Floroglucinol/análogos & derivados , Terpenos/farmacologia , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Glutationa Peroxidase/metabolismo , Humanos , Malondialdeído/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Compostos Organometálicos/toxicidade , Células PC12 , Floroglucinol/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pironas/toxicidade , Ratos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
11.
Chemosphere ; 209: 972-980, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30114748

RESUMO

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.


Assuntos
Inflamassomos/metabolismo , Interleucina-1beta/genética , Lipopolissacarídeos/metabolismo , Macrófagos Peritoneais/metabolismo , Inibidor de NF-kappaB alfa/genética , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , Transdução de Sinais
12.
Chemosphere ; 203: 170-178, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29614410

RESUMO

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.


Assuntos
Compostos de Alumínio/toxicidade , Cloretos/toxicidade , Hipocampo/patologia , Interleucina-1beta/metabolismo , Transdução de Sinais , Alumínio/metabolismo , Cloreto de Alumínio , Animais , Citocinas/metabolismo , Hipocampo/metabolismo , Inflamação/induzido quimicamente , Neurônios/metabolismo , Ratos , Ratos Wistar
13.
Biol Trace Elem Res ; 184(1): 173-185, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29027106

RESUMO

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.


Assuntos
Compostos de Alumínio/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cloretos/farmacologia , RNA Mensageiro/metabolismo , Receptor fas/metabolismo , Cloreto de Alumínio , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Proteína Ligante Fas/metabolismo , Citometria de Fluxo , Mitocôndrias/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos
14.
J Inorg Biochem ; 174: 55-62, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28605655

RESUMO

Aluminum (Al) is known to exert hepatotoxicity. However, the mechanisms mostly are unclear. Liver is a metabolism organ that maintains the energy level and structural stability of body, mitochondria are the main sites of energy metabolism, thus, we hypothesized that mitochondrial energy metabolism disorder contributes to liver dysfunction in aluminum chloride (AlCl3) treatment rat. To verify the hypothesis, forty male Wistar rats were randomly allocated and orally exposed to 0, 64mg/kg, 128mg/kg and 256mg/kg body weight AlCl3 in drinking water for 120days, respectively. We found that AlCl3 exposure reduced the electron transport chain complexes I-V activities and adenosine triphosphate (ATP) level, as well as disturbed mitochondrial DNA transcript, presenting as the inhibited mRNA expressions of NADH dehydrogenase 1, NADH dehydrogenase 2, cytochrome b, cytochrome c oxidase subunit 1, cytochrome c oxidase subunit 3 and ATP synthase 6, indicating that AlCl3 exposure disturbs the mitochondrial energy metabolism, and it caused an increase in liver enzymes (Aspartate aminotransferase and Alanine aminotransferase) and histopathological lesions. Additionally, we found that reactive oxygen species accumulation and decreased superoxide dismutase activity in mitochondria, and increased 8-Hydroxydeoxyguanosine levels in mitochondrial DNA, demonstrating AlCl3 exposure promotes mitochondrial oxidative stress, which may be a contributing factor to mitochondrial energy metabolism disorder and liver dysfunction. The study displayed that mitochondria are the potential target of liver damage induced by AlCl3, providing considerable direction for the prevention and clinical intervention of liver diseases.


Assuntos
Compostos de Alumínio/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Cloretos/toxicidade , Metabolismo Energético/efeitos dos fármacos , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , Cloreto de Alumínio , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fígado/patologia , Masculino , Mitocôndrias Hepáticas/patologia , Ratos , Ratos Wistar
15.
Biol Trace Elem Res ; 180(2): 233-238, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28349382

RESUMO

Aflatoxins have been considered as one of the major risk factors of male infertility, and aflatoxin B1 (AFB1) is the most highly toxic and prevalent member of the aflatoxins family. Selenium (Se), an essential nutritional trace mineral for normal testicular development and male fertility, has received extensive intensive on protective effects of male reproductive system due to its potential antioxidant and activating testosterone synthesis. To investigate the protective effect of Se on AFB1-induced testicular toxicity, the mice were orally administered with AFB1 (0.75 mg/kg) and Se (0.2 mg/kg or 0.4 mg/kg) for 45 days. We found that that Se elevated testes index, sperm functional parameters (concentration, malformation, and motility), and the level of serum testosterone in AFB1-exposed mice. Moreover, our results showed that Se attenuated the AFB1-induced oxidative stress and the reduction of testicular testosterone synthesis enzyme protein expression such as steroidogenic acute regulatory protein (StAR), P450 side-chain cleavage (P450scc), and 17ß-hydroxysteroid dehydrogenase (17ß-HSD) in AFB1-exposed mice. These results demonstrated that Se conferred protection against AFB1-induced testicular toxicity and can be attributed to its antioxidant and increased testosterone level by stimulating protein expression of StAR and testosterone synthetic enzymes.


Assuntos
Aflatoxina B1/antagonistas & inibidores , Suplementos Nutricionais , Infertilidade Masculina/prevenção & controle , Estresse Oxidativo , Substâncias Protetoras/uso terapêutico , Selênio/uso terapêutico , Testículo/efeitos dos fármacos , 17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Aflatoxina B1/toxicidade , Animais , Animais não Endogâmicos , Antioxidantes/uso terapêutico , Biomarcadores/sangue , Biomarcadores/metabolismo , Carcinógenos Ambientais/química , Carcinógenos Ambientais/toxicidade , Enzima de Clivagem da Cadeia Lateral do Colesterol/antagonistas & inibidores , Enzima de Clivagem da Cadeia Lateral do Colesterol/química , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Contaminação de Alimentos , Doenças Transmitidas por Alimentos/etiologia , Doenças Transmitidas por Alimentos/prevenção & controle , Infertilidade Masculina/sangue , Infertilidade Masculina/induzido quimicamente , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Fosfoproteínas/agonistas , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/metabolismo , Substâncias Protetoras/administração & dosagem , Selênio/administração & dosagem , Análise do Sêmen , Selenito de Sódio/administração & dosagem , Testículo/metabolismo , Testosterona/biossíntese , Testosterona/sangue
16.
Environ Toxicol Pharmacol ; 48: 125-129, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27771505

RESUMO

Aluminum (Al) exposure impairs bone formation, and bone formation is mediated by the osteoblasts. But effects of Al on the osteoblasts function remain elusive. The osteoblasts were exposed to 0, 0.0252, 0.126, 0.252mg/mL AlCl3·6H2O for 24h. The osteoblasts viability, TGF-ß1, BMP-2, IGF-I and Cbfα1 mRNA expressions, and GSH-Px and SOD activities, ROS concentration were determined. The osteoblasts ultrastructural features were also observed. The results showed that AlCl3 suppressed the osteoblasts viability, TGF-ß1, BMP-2, IGF-I and Cbfα1 mRNA expressions, GSH-Px and SOD activities, and elevated ROS concentration compared with the CG. The ultrastructural features of osteoblasts in the HG showed mitochondrial swelling, foam-like structure, uneven distribution of chromatin, incomplete cell membrane and cytoplasm spillover compared with the CG. It indicates that AlCl3 inhibits osteoblasts viability, growth regulation factors mRNA expressions, anti-oxidative function, and damaged the osteoblasts histology structure, impairing the osteoblasts function.


Assuntos
Compostos de Alumínio/toxicidade , Cloretos/toxicidade , Osteoblastos/efeitos dos fármacos , Cloreto de Alumínio , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Dilatação Mitocondrial/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
17.
Chemosphere ; 151: 289-95, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26946116

RESUMO

Aluminum (Al) is present in the daily life of humans, and the incidence of Al contamination increased in recent years. Long-term excessive Al intake induces neuroinflammation and cognition impairment. Neuroinflammation alter density of dendritic spine, which, in turn, influence cognition function. However, it is unknown whether increased neuroinflammation is associated with altered density of dendritic spine in Al-treated rats. In the present study, AlCl3 was orally administrated to rat at 50, 150 and 450 mg/kg for 90d. We examined the effects of AlCl3 on the cognition function, density of dendritic spine in hippocampus of CA1 and DG region and the mRNA levels of IL-1ß, IL-6, TNF-α, MHC II, CX3CL1 and BNDF in developing rat. These results showed exposure to AlCl3 lead to increased mRNA levels of IL-1ß, IL-6, TNF-α and MCH II, decreased mRNA levels of CX3CL1 and BDNF, decreased density of dendritic spine and impaired learning and memory in developing rat. Our results suggest AlCl3 can induce neuroinflammation that may result in loss of spine, and thereby leads to learning and memory deficits.


Assuntos
Compostos de Alumínio/toxicidade , Cloretos/toxicidade , Transtornos Cognitivos/induzido quimicamente , Cognição/efeitos dos fármacos , Citocinas/imunologia , Espinhas Dendríticas/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Administração Oral , Cloreto de Alumínio , Animais , Transtornos Cognitivos/imunologia , Transtornos Cognitivos/patologia , Citocinas/genética , Espinhas Dendríticas/imunologia , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hipocampo/imunologia , Hipocampo/patologia , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
18.
Biol Trace Elem Res ; 173(2): 399-404, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27008427

RESUMO

Aluminum (Al) exposure is toxic to immune system. Studies have implicated that glucocorticoids (GCs) exert the dual regulation effect on the immune function depending on the concentration. However, it is unknown whether a dual effect of GCs exists in the AlCl3-treated lymphocytes. Corticosterone (Cort) is one kind of GCs. To investigate the effect of different concentration Cort on AlCl3-treated lymphocytes, rat splenic lymphocyte was isolated and cultured with 0.55 mmol/L AlCl3, simultaneously administrated Cort at final concentration of 0 (control group, CG), 10(-8) (low-level group, LG), and 10(-6) (high-level group, HG) mol/L, respectively. Another group without AlCl3 and Cort served as the blank group (BG). We found that low concentration Cort increased the T and B lymphocyte proliferation rate, proportions of CD4(+) T lymphocyte subset, IgG, IL-2, and TNF-α contents, whereas high concentration Cort decreased those in AlCl3-treated lymphocytes. In conclusion, the results of this study indicated that low concentration Cort relieves the immunotoxicity of AlCl3 on the splenic lymphocytes, whereas high concentration Cort aggravates it.


Assuntos
Compostos de Alumínio/toxicidade , Cloretos/toxicidade , Corticosterona/farmacologia , Linfócitos/imunologia , Baço/imunologia , Cloreto de Alumínio , Animais , Células Cultivadas , Linfócitos/citologia , Masculino , Ratos , Ratos Wistar , Baço/citologia
19.
Biol Trace Elem Res ; 173(2): 339-44, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26910335

RESUMO

This study investigated the toxicity of aluminum chloride (AlCl3) exposure in the rat kidney. Forty male Wistar rats (5 weeks old), weighing 110-120 g, were randomly divided into four groups: control group (CG, 0 g/L AlCl3), low dose group (LG, 0.4 g/L AlCl3), mid dose group (MG, 0.8 g/L AlCl3), and high dose group (HG, 1.6 g/L AlCl3). Rats were administered AlCl3 in their drinking water for 120 days. A variety of measurements were taken including superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) activities, malondialdehyde (MDA) concentration in the kidney and blood urea nitrogen (BUN), and cystatin C (Cys-C) concentrations in the serum. In addition, Al and ß2-microglobulin (ß2-MG) concentrations and the activity of N-acetyl-ß-D-glucosaminidase (NAG) in the urine were determined. The results showed that in the AlCl3-treated groups SOD and GSH-PX activities were decreased, while NAG activity and Al, MDA, BUN, Cys-C, and ß2-MG concentrations were increased, compared with the CG. This study indicates that AlCl3 exposure induces oxidative stress and suppresses kidney function.


Assuntos
Compostos de Alumínio/toxicidade , Cloretos/toxicidade , Rim/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Cloreto de Alumínio , Animais , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/biossíntese , Rim/patologia , Masculino , Malondialdeído/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/biossíntese
20.
Environ Toxicol Pharmacol ; 42: 198-204, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26878280

RESUMO

Exposure to aluminum (Al) suppresses bone formation. Osteoblastic differentiation plays a key role in the process of bone formation. However, the effect of Al on osteoblastic differentiation is still controversial, and the mechanism remains unclear. To investigate the effect of Al on osteoblastic differentiation and whether Wnt signaling pathway was involved in it, the primary rat osteoblasts were exposed to 1/40 IC50, 1/20 IC50 and 1/10 IC50 of aluminum trichloride (AlCl3) for 24h, respectively. The activity analysis of alkaline phosphate, qRT-PCR analysis of type I collagen, alkaline phosphate, Wnt3a and Dkk-1, Western blot analysis of p-GSK3ß, GSK3ß and ß-catenin protein and Immunofluorescence staining for ß-catenin suggested that AlCl3 inhibited osteoblastic differentiation and Wnt/ß-catenin pathway. Moreover, we found exogenous Wnt3a application reversed the inhibitory effect of AlCl3 on osteoblastic differentiation, accompanied by activating the Wnt/ß-catenin pathway. Taken together, these findings suggest that AlCl3 inhibites osteoblastic differentiation through inactivation of Wnt/ß-catenin pathway in osteoblasts.


Assuntos
Compostos de Alumínio/toxicidade , Cloretos/toxicidade , Substâncias Perigosas/toxicidade , Osteoblastos/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Cloreto de Alumínio , Animais , Diferenciação Celular/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Osteogênese/efeitos dos fármacos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA