Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.484
Filtrar
1.
Ecotoxicol Environ Saf ; 281: 116661, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38954907

RESUMO

OBJECTIVE: Baicalin has antioxidative, antiviral, and anti-inflammatory properties. However, its ability to alleviate oxidative stress (OS) and DNA damage in liver cells exposed to aflatoxin B1 (AFB1), a highly hepatotoxic compound, remains uncertain. In this study, the protective effects of baicalin on AFB1-induced hepatocyte injury and the mechanisms underlying those effects were investigated. METHODS: Stable cell lines expressing CYP3A4 were established using lentiviral vectors to assess oxidative stress levels by conducting assays to determine the content of reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD). Additionally, DNA damage was evaluated by 8-hydroxy-2-deoxyguanosine (8-OHdG) and comet assays. Transcriptome sequencing, molecular docking, and in vitro experiments were conducted to determine the mechanisms underlying the effects of baicalin on AFB1-induced hepatocyte injury. In vivo, a rat model of hepatocyte injury induced by AFB1 was used to evaluate the effects of baicalin. RESULTS: In vitro, baicalin significantly attenuated AFB1-induced injury caused due to OS, as determined by a decrease in ROS, MDA, and SOD levels. Baicalin also considerably decreased AFB1-induced DNA damage in hepatocytes. This protective effect of baicalin was found to be closely associated with the TP53-mediated ferroptosis pathway. To elaborate, baicalin physically interacts with P53, leading to the suppression of the expression of GPX4 and SLC7A11, which in turn inhibits ferroptosis. In vivo findings showed that baicalin decreased DNA damage and ferroptosis in AFB1-treated rat liver tissues, as determined by a decrease in the expression of γ-H2AX and an increase in GPX4 and SLC7A11 levels. Overexpression of TP53 weakened the protective effects of baicalin. CONCLUSIONS: Baicalin can alleviate AFB1-induced OS and DNA damage in liver cells via the TP53-mediated ferroptosis pathway. In this study, a theoretical foundation was established for the use of baicalin in protecting the liver from the toxic effects of AFB1.


Assuntos
Aflatoxina B1 , Ferroptose , Flavonoides , Hepatócitos , Proteína Supressora de Tumor p53 , Flavonoides/farmacologia , Aflatoxina B1/toxicidade , Ferroptose/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Animais , Proteína Supressora de Tumor p53/metabolismo , Ratos , Estresse Oxidativo/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Masculino , Substâncias Protetoras/farmacologia , Ratos Sprague-Dawley , Humanos , Espécies Reativas de Oxigênio/metabolismo
3.
J Hazard Mater ; 474: 134822, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38850943

RESUMO

The disturbed gut microbiota is a key factor in activating the aflatoxin B1 (AFB1)-induced liver pyroptosis by promoting inflammatory hepatic injury; however, the pathogen associated molecular pattern (PAMP) from disturbed gut microbiota and its mechanism in activating liver pyroptosis remain undefined. By transplanting AFB1-originated fecal microbiota and sterile fecal microbial metabolites filtrate, we determined the association of PAMP in AFB1-induced liver pyroptosis. Notably, AFB1-originated sterile fecal microbial metabolites filtrate were more active in triggering liver pyroptosis in mice, as compared to parental fecal microbiota. This result supported a critical role of the metabolic homeostasis of gut microbiota in AFB1-induced liver pyroptosis, rather than an injurious response to direct exposure of AFB1 in liver. Among the gut-microbial metabolites, pipecolic acid and norepinephrine were proposed to bind TLR4 and NLRP3, the upstream proteins of pyroptosis signaling pathway. Besides, the activations of TLR4 and NLRP3 were linearly correlated with the concentrations of pipecolic acid and norepinephrine in the serum of mice. In silenced expression of TLR4 and NLRP3 in HepG2 cells, pipecolic acid or norepinephrine did not able to activate hepatocyte pyroptosis. These results demonstrated the necessity of gut microbial metabolism in sustaining liver homeostasis, as well as the potential to provide new insights into targeted intervention for AFB1 hepatotoxicity.


Assuntos
Aflatoxina B1 , Microbioma Gastrointestinal , Fígado , Proteína 3 que Contém Domínio de Pirina da Família NLR , Norepinefrina , Ácidos Pipecólicos , Piroptose , Animais , Aflatoxina B1/toxicidade , Aflatoxina B1/metabolismo , Piroptose/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Ácidos Pipecólicos/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Norepinefrina/metabolismo , Células Hep G2 , Masculino , Camundongos Endogâmicos C57BL , Receptor 4 Toll-Like/metabolismo , Camundongos , Fezes/microbiologia
4.
Pestic Biochem Physiol ; 202: 105944, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38879301

RESUMO

Contamination of food products with mycotoxins such as aflatoxin B1 (AFB1) poses a severe risk to human health. Larvae of the black soldier fly (BSFL), Hermetia illucens (Diptera: Stratiomyidae), can successfully metabolize AFB1 without any negative consequences on their survival or growth. Organic waste streams contaminated with mycotoxins can be upcycled into protein-rich BSFL as an alternative feed for livestock and the left-over feed residue into nutrient-rich crop fertilizers. However, the underlying mechanisms that allow BSFL to metabolize AFB1 are unknown. In this study, five-day-old BSFL were fed with either a control or an AFB1-spiked (20 µg/kg) diet to elucidate the underlying mechanisms. Larval samples were collected at three timepoints (6 h, 24 h and 72 h) and subjected to RNA-Seq analysis to determine gene expression patterns. Provision of an AFB1-spiked diet resulted in an up-regulation of 357 and a down-regulation of 929 unique genes. Upregulated genes include multiple genes involved in AFB1 metabolism in other (insect) species. Downregulated genes were generally involved in the insects' growth, development, and immunity. BSFL possesses a diverse genetic arsenal that encodes for enzymes capable of metabolizing AFB1 without trade-offs on larval survival. In conclusion, the adverse impact of AFB1 exposure on immunity-related processes is observed in the transcriptomic response, and is indicative of a trade-off between detoxification and immune responses.


Assuntos
Aflatoxina B1 , Dípteros , Larva , Animais , Aflatoxina B1/toxicidade , Dípteros/efeitos dos fármacos , Dípteros/genética , Dípteros/metabolismo , Larva/efeitos dos fármacos , Larva/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos
5.
Ecotoxicol Environ Saf ; 281: 116619, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38925031

RESUMO

This study investigated the effects of compound probiotics (CP) on AFB1-induced cytotoxicity in Sertoli TM4 cells. The L9 (3 × 3) orthogonal test was conducted to determine the optimal CP required for high AFB1 degradation in the artificial gastrointestinal fluid in vitro. The maximal AFB1 degradation rate was 40.55 % (P < 0.05) when the final viable count was 1.0 × 105 CFU/mL for Bacillus subtilis, Lactobacillus casein, and Saccharomyces cerevisiae. The effects of CP and the CP supernatant (CPS) on TM4 cell viability were evaluated to achieve the optimal protective conditions. When CPS4 (corresponding to CP viable counts of 1.0 × 104 CFU/mL) was added to the TM4 cells for 24 h, the cell viability reached 108.86 % (P < 0.05). AFB1 reduced TM4 cell viability in a concentration- and time-dependent manner at an AFB1 concentration ranging from 0 to 1.5 µM after 48-h AFB1 exposure. The optimal AFB1 concentration/times for low- and high damage models were 0.5 and 1.25 µM both for 24 h, which decreased viability to 76.04 % and 65.35 %, respectively. however, CPS4 added to low- and high-damage models increased the cell viability to 97.43 % and 75.12 %, respectively (P < 0.05). Transcriptome sequencing was performed based on the following designed groups: the control, 0.5 µM AFB1, 1.25 µM AFB1, CPS4, and CPS4+0.5 µM AFB1. The Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis was further performed to identify significantly enriched signaling pathways, which were subsequently verified. It was shown that AFB1 induced apoptosis by blocking the PI3K-AKT-mTOR pathway and upregulating autophagy proteins such as LC3B, Beclin1, and ATG5 while inhibiting autophagic flux. CPS4 promoted AFB1 degradation, activated the p62-NRF2 antioxidant, and inhibited ROS/TRPML1 pathways, thereby reducing ROS production and inflammation and ultimately alleviating AFB1-induced autophagy and apoptosis. These findings supports the potential of probiotics to protect the male reproductive system from toxin damage.


Assuntos
Aflatoxina B1 , Antioxidantes , Autofagia , Sobrevivência Celular , Fator 2 Relacionado a NF-E2 , Probióticos , Células de Sertoli , Probióticos/farmacologia , Animais , Aflatoxina B1/toxicidade , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Autofagia/efeitos dos fármacos , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Masculino , Sobrevivência Celular/efeitos dos fármacos , Linhagem Celular , Transdução de Sinais/efeitos dos fármacos
6.
Ecotoxicol Environ Saf ; 280: 116527, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38833978

RESUMO

Aflatoxin B1 (AFB1) is known to inhibit growth, and inflict hepatic damage by interfering with protein synthesis. Allicin, has been acknowledged as an efficacious antioxidant capable of shielding the liver from oxidative harm. This study aimed to examine the damage caused by AFB1 on bovine hepatic cells and the protective role of allicin against AFB1-induced cytotoxicity. In this study, cells were pretreated with allicin before the addition of AFB1 for co-cultivation. Our findings indicate that AFB1 compromises cellular integrity, suppresses the expression of nuclear factor erythroid 2-related factor 2 (Nrf2). In addition, allicin attenuates oxidative damage to bovine hepatic cells caused by AFB1 by promoting the expression of the Nrf2 pathway and reducing cell apoptosis. In conclusion, the results of this study will help advance clinical research and applications, providing new options and directions for the prevention and treatment of liver diseases.


Assuntos
Aflatoxina B1 , Antioxidantes , Apoptose , Dissulfetos , Hepatócitos , Fator 2 Relacionado a NF-E2 , Estresse Oxidativo , Transdução de Sinais , Ácidos Sulfínicos , Animais , Ácidos Sulfínicos/farmacologia , Aflatoxina B1/toxicidade , Bovinos , Dissulfetos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Antioxidantes/farmacologia , Feminino
7.
Ecotoxicol Environ Saf ; 280: 116574, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38875822

RESUMO

Aflatoxin B1 (AFB1) is commonly found in feed ingredients and foods all over the world, posing a significant threat to food safety and public health in animals and humans. Lactobacillus salivarius (L. salivarius) was recorded to improve the intestinal health and performance of chickens. However, whether L. salivarius can alleviate AFB1-induced hepatotoxicity in geese was unknown. A total of 300 Lande geese were randomly assigned to five groups: control group, AFB1 low-dose group (L), L. salivarius+AFB1 low-dose group (LL), AFB1 high dosage groups (H), L. salivarius+AFB1 high dosage groups (LH), respectively. The results showed that the concentrations of ALT, AST, and GGT significantly increased after exposure to AFB1. Similarly, severe damage of hepatic morphology was observed including the hepatic structure injury and inflammatory cell infiltration. The oxidative stress was evidenced by the elevated concentrations of MDA, and decreased activities of GSH-Px, GSH and SOD. The observation of immunofluorescence, real-time PCR, and western blotting showed that the expression of PINK1 and the value of LC3II/LC3I were increased, but that of p62 significantly decreased after AFB1 exposure. Moreover, the supplementation of L. salivarius effectively improved the geese performance, ameliorated AFB1-induced oxidative stress, inhibited mitochondrial mitophagy and enhanced the liver restoration to normal level. The present study demonstrated that L. salivarius ameliorated AFB1-induced the hepatotoxicity by decreasing the oxidative stress, and regulating the expression of PINK1/Parkin-mediated mitophagy in the mitochondria of the geese liver. Furthermore, this investigation suggested that L. salivarius might serve as a novel and safe additive for preventing AFB1 contamination in poultry feed.


Assuntos
Aflatoxina B1 , Gansos , Ligilactobacillus salivarius , Fígado , Mitofagia , Proteínas Quinases , Ubiquitina-Proteína Ligases , Animais , Aflatoxina B1/toxicidade , Mitofagia/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Ligilactobacillus salivarius/fisiologia , Fígado/efeitos dos fármacos , Fígado/patologia , Proteínas Quinases/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Doença Hepática Induzida por Substâncias e Drogas/patologia , Estresse Oxidativo/efeitos dos fármacos , Probióticos/farmacologia
8.
Mycotoxin Res ; 40(3): 457-466, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38913091

RESUMO

Aflatoxin B1 (AFB1) is classified as a Class I carcinogen and common pollutant in human and animal food products. Prolonged exposure to AFB1 can induce hepatocyte apoptosis and lead to hepatotoxicity. Therefore, preventing AFB1-induced hepatotoxicity remains a critical issue and is of great significance. Baicalin, a polyphenolic compound derived from Scutellaria baicalensis Georgi, has a variety of pharmacodynamic activities, such as antiapoptotic and anticancer activities. This study systematically investigated the alleviating effect of baicalin on AFB1-induced hepatotoxicity from the perspective of apoptosis and explored the possible molecular mechanism. In the normal human liver cell line L02, baicalin treatment significantly inhibited AFB1-induced c-Jun-N-terminal Kinase (JNK) activation and cell apoptosis. In addition, the in vitro mechanism study demonstrated that baicalin alleviates AFB1-induced hepatocyte apoptosis through suppressing the translocation of phosphorylated JNK to the nucleus and decreasing the phosphorylated c-Jun/c-Jun ratio and the Bax/Bcl2 ratio. Molecular docking and drug affinity responsive target stability assays demonstrated that baicalin has the potential to target JNK. This study provides a basis for the therapeutic effect of baicalin on hepatocyte apoptosis caused by AFB1, indicating that the development of baicalin and JNK pathway inhibitors has broad application prospects in the prevention of hepatotoxicity, especially hepatocyte apoptosis.


Assuntos
Aflatoxina B1 , Apoptose , Flavonoides , Hepatócitos , Proteínas Quinases JNK Ativadas por Mitógeno , Flavonoides/farmacologia , Apoptose/efeitos dos fármacos , Aflatoxina B1/toxicidade , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Hepatócitos/efeitos dos fármacos , Linhagem Celular , Simulação de Acoplamento Molecular , Scutellaria baicalensis/química
9.
Toxins (Basel) ; 16(6)2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38922135

RESUMO

The aim of this study was to investigate the effects of aflatoxin B1 (AFB1) on cholestasis in duck liver and its nutritional regulation. Three hundred sixty 1-day-old ducks were randomly divided into six groups and fed for 4 weeks. The control group was fed a basic diet, while the experimental group diet contained 90 µg/kg of AFB1. Cholestyramine, atorvastatin calcium, taurine, and emodin were added to the diets of four experimental groups. The results show that in the AFB1 group, the growth properties, total bile acid (TBA) serum levels and total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), and glutathione (GSH) liver levels decreased, while the malondialdehyde (MDA) and TBA liver levels increased (p < 0.05). Moreover, AFB1 caused cholestasis. Cholestyramine, atorvastatin calcium, taurine, and emodin could reduce the TBA serum and liver levels (p < 0.05), alleviating the symptoms of cholestasis. The qPCR results show that AFB1 upregulated cytochrome P450 family 7 subfamily A member 1 (CYP7A1) and cytochrome P450 family 8 subfamily B member 1 (CYP8B1) gene expression and downregulated ATP binding cassette subfamily B member 11 (BSEP) gene expression in the liver, and taurine and emodin downregulated CYP7A1 and CYP8B1 gene expression (p < 0.05). In summary, AFB1 negatively affects health and alters the expression of genes related to liver bile acid metabolism, leading to cholestasis. Cholestyramine, atorvastatin calcium, taurine, and emodin can alleviate AFB1-induced cholestasis.


Assuntos
Aflatoxina B1 , Colestase , Patos , Fígado , Animais , Aflatoxina B1/toxicidade , Colestase/induzido quimicamente , Colestase/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/sangue , Doenças das Aves Domésticas/induzido quimicamente , Resina de Colestiramina/farmacologia , Ração Animal
10.
Toxins (Basel) ; 16(6)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38922153

RESUMO

Physiologically based pharmacokinetic (PBPK) models were utilized to investigate potential interactions between aflatoxin B1 (AFB1) and efavirenz (EFV), a non-nucleoside reverse transcriptase inhibitor drug and inducer of several CYP enzymes, including CYP3A4. PBPK simulations were conducted in a North European Caucasian and Black South African population, considering different dosing scenarios. The simulations predicted the impact of EFV on AFB1 metabolism via CYP3A4 and CYP1A2. In vitro experiments using human liver microsomes (HLM) were performed to verify the PBPK predictions for both single- and multiple-dose exposures to EFV. Results showed no significant difference in the formation of AFB1 metabolites when combined with EFV (0.15 µM) compared to AFB1 alone. However, exposure to 5 µM of EFV, mimicking chronic exposure, resulted in increased CYP3A4 activity, affecting metabolite formation. While co-incubation with EFV reduced the formation of certain AFB1 metabolites, other outcomes varied and could not be fully attributed to CYP3A4 induction. Overall, this study provides evidence that EFV, and potentially other CYP1A2/CYP3A4 perpetrators, can impact AFB1 metabolism, leading to altered exposure to toxic metabolites. The results emphasize the importance of considering drug interactions when assessing the risks associated with mycotoxin exposure in individuals undergoing HIV therapy in a European and African context.


Assuntos
Aflatoxina B1 , Alcinos , Benzoxazinas , Ciclopropanos , Interações Medicamentosas , Microssomos Hepáticos , Modelos Biológicos , Inibidores da Transcriptase Reversa , Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidade , Humanos , Benzoxazinas/farmacocinética , Benzoxazinas/metabolismo , Microssomos Hepáticos/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Inibidores da Transcriptase Reversa/farmacocinética , Masculino , Citocromo P-450 CYP3A/metabolismo , Adulto , Feminino , Citocromo P-450 CYP1A2/metabolismo , Pessoa de Meia-Idade , Adulto Jovem , População Branca
11.
Int J Biol Macromol ; 271(Pt 1): 132371, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38750861

RESUMO

Aflatoxin B1 (AFB1) is one of the most widespread contaminants in agricultural commodities. Pleurotus eryngii (PE) is widely used as a feed additive for its anti-inflammatory properties, and its major active substance is believed to be polysaccharides. This study aims to explore the underlying mechanism of dietary PE polysaccharides alleviating AFB1-induced toxicity in ducks. The major monosaccharide components of PE polysaccharides were identified as glucose, mannose, galactose, glucuronic acid, and fucose. The results showed that dietary PE polysaccharides could alleviate liver inflammation, alleviate intestinal barrier dysfunction, and change the imbalanced gut microbiota induced by AFB1 in ducks. However, PE polysaccharides failed to exert protective roles on the liver and intestine injury induced by AFB1 in antibiotic-treated ducks. The PE + AFB1-originated microbiota showed a positive effect on intestinal barrier and inflammation, the SCFAs transport via the gut-liver axis, and liver inflammation compared with the AFB1-originated microbiota in ducks. These findings provided a possible mechanism that PE polysaccharides alleviated AFB1-induced liver inflammation in ducks by remodeling gut microbiota, regulating microbiota-derived SCFAs transport via the gut-liver axis, and inhibiting inflammatory gene expressions in the liver, which may provide new insight for therapeutic methods against AFB1 exposure in animals.


Assuntos
Aflatoxina B1 , Patos , Microbioma Gastrointestinal , Fígado , Pleurotus , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Aflatoxina B1/toxicidade , Pleurotus/química , Fígado/efeitos dos fármacos , Fígado/metabolismo , Polissacarídeos/farmacologia , Polissacarídeos/química , Ácidos Graxos Voláteis/metabolismo , Polissacarídeos Fúngicos/farmacologia , Polissacarídeos Fúngicos/química , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/induzido quimicamente , Transporte Biológico/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico
12.
Food Chem Toxicol ; 189: 114745, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38763499

RESUMO

Mycotoxins are toxic metabolites produced by fungal species, commonly exist in animal feeds, and pose a serious risk to human as well as animal health. But limited studies have focused on combined effects of no-observed adverse effect levels. In vivo study, 6 weeks old twenty-four mice were individually exposed to Deoxynivalenol (DON) at 0.1 mg/kg BW, Aflatoxin B1 (AFB1) at 0.01 mg/kg BW, and mixture of DON and AFB1 (0.1 mg/kg BW and 0.01 mg/kg BW, respectively) for 28 days. Then, DON at 0.5 µg/mL, AFB1 at 0.04 µg/mL, and mixtures of DON and AFB1 (0.5 µg/mL, 0.04 µg/mL, respectively) were applied to porcine alveolar macrophages (PAMs) in vitro study. Our in vivo results revealed that the combined no-observed adverse effect levels of DON and AFB1 administration decreased IgA and IgG levels in the serum, the splenic TNF-α, IFN-γ, IL-2 and IL-6 mRNA expression and T-lymphocyte subset levels (CD4+ and CD8+) in the spleen. Additionally, the combined administration increased caspase-3, caspase-9, Bax, Cyt-c, and decreased Bcl-2 protein expression. Taken together, the combined no-observed adverse effect levels of DON and AFB1 could induce immunosuppression, which may be related to apoptosis. This study provides new insights into the combined immune toxicity (DON and AFB1).


Assuntos
Aflatoxina B1 , Apoptose , Tricotecenos , Animais , Tricotecenos/toxicidade , Aflatoxina B1/toxicidade , Apoptose/efeitos dos fármacos , Camundongos , Suínos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Baço/efeitos dos fármacos , Masculino , Citocinas/metabolismo , Imunoglobulina A , Feminino
13.
J Neuroimmunol ; 391: 578365, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38723577

RESUMO

Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficiencies in communication, repetitive and stereotyped behavioral patterns, and difficulties in reciprocal social engagement. The presence of immunological dysfunction in ASD has been well established. Aflatoxin B1 (AFB1) is a prevalent mycotoxin found in food and feed, causing immune toxicity and hepatotoxicity. AFB1 is significantly elevated in several regions around the globe. Existing research indicates that prolonged exposure to AFB1 results in neurological problems. The BTBR T+ Itpr3tf/J (BTBR) mice, which were used as an autism model, exhibit the primary behavioral traits that define ASD, such as repeated, stereotyped behaviors and impaired social interactions. The main objective of this work was to assess the toxic impact of AFB1 in BTBR mice. This work aimed to examine the effects of AFB1 on the expression of Notch-1, IL-6, MCP-1, iNOS, GM-CSF, and NF-κB p65 by CD19+ B cells in the spleen of the BTBR using flow cytometry. We also verified the impact of AFB1 exposure on the mRNA expression levels of Notch-1, IL-6, MCP-1, iNOS, GM-CSF, and NF-κB p65 in the brain of BTBR mice using real-time PCR. The findings of our study showed that the mice treated with AFB1 in the BTBR group exhibited a substantial increase in the presence of CD19+Notch-1+, CD19+IL-6+, CD19+MCP-1+, CD19+iNOS+, CD19+GM-CSF+, and CD19+NF-κB p65+ compared to the mice in the BTBR group that were treated with saline. Our findings also confirmed that administering AFB1 to BTBR mice leads to elevated mRNA expression levels of Notch-1, IL-6, MCP-1, iNOS, GM-CSF, and NF-κB p65 in the brain, in comparison to BTBR mice treated with saline. The data highlight that exposure to AFB1 worsens immunological abnormalities by increasing the expression of inflammatory mediators in BTBR mice.


Assuntos
Aflatoxina B1 , Antígenos CD19 , Modelos Animais de Doenças , Animais , Camundongos , Aflatoxina B1/toxicidade , Antígenos CD19/metabolismo , Masculino , Mediadores da Inflamação/metabolismo , Transtorno Autístico/induzido quimicamente , Transtorno Autístico/imunologia , Transtorno Autístico/metabolismo , Camundongos Transgênicos
14.
Basic Clin Pharmacol Toxicol ; 135(2): 195-209, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38804152

RESUMO

Aflatoxin B1 (AFB1) is extremely harmful to both humans and animals. Mitophagy is a selective process of self-elimination and has an important role in controlling mitochondrial quality. The present study aimed to investigate the effect of reactive oxygen species (ROS) accumulation on AFB1-induced mitophagy in HepG2 cells to provide a new perspective from which to design novel therapeutic strategies to treat AFB1 poisoning. ROS release was induced in HepG2 cells with AFB1 (10 µmol/L). Cell autophagy activity, mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) levels, Parkin translocation and both the transcription and expression of mitophagy-related proteins were measured when N-acetyl-L-cysteine (NAC) partially decreased the ROS level, while the knockdown of nuclear factor erythroid 2-related factor 2 (Nrf2) resulted in a large accumulation of ROS. The results reveal that NAC pretreatment ameliorated the decline in both the MMP and the ATP levels while also activating phosphoglycerate mutase 5 (PGAM5)-PTEN-induced kinase 1 (PINK1)/Parkin, while the Nrf2 knockdown group exhibited the opposite trend. These results suggest that AFB1-induced mitophagy in HepG2 cells depends on ROS, and proper ROS activates mitophagy to play a protective role.


Assuntos
Trifosfato de Adenosina , Aflatoxina B1 , Potencial da Membrana Mitocondrial , Mitofagia , Fator 2 Relacionado a NF-E2 , Proteínas Quinases , Espécies Reativas de Oxigênio , Ubiquitina-Proteína Ligases , Humanos , Mitofagia/efeitos dos fármacos , Células Hep G2 , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Quinases/metabolismo , Aflatoxina B1/toxicidade , Trifosfato de Adenosina/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais/efeitos dos fármacos , Fosfoproteínas Fosfatases/metabolismo , Fosfoproteínas Fosfatases/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Acetilcisteína/farmacologia , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética
15.
Toxins (Basel) ; 16(5)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38787059

RESUMO

The aims of this study were (i) to determine the effect of an algoclay-based decontaminant on the oral availability of three mycotoxins (deoxynivalenol; DON, ochratoxin A; OTA, and aflatoxin B1; AFB1) using an oral bolus model and (ii) to determine the effect of this decontaminant on the performance, intestinal morphology, liver oxidative stress, and metabolism, in broiler chickens fed a diet naturally contaminated with DON. In experiment 1, sixteen 27-day-old male chickens (approximately 1.6 kg body weight; BW) were fasted for 12 h and then given a bolus containing either the mycotoxins (0.5 mg DON/kg BW, 0.25 mg OTA/kg BW, and 2.0 mg AFB1/kg BW) alone (n = 8) or combined with the decontaminant (2.5 g decontaminant/kg feed; circa 240 mg/kg BW) (n = 8). Blood samples were taken between 0 h (before bolus administration) and 24 h post-administration for DON-3-sulphate, OTA, and AFB1 quantification in plasma. The algoclay decontaminant decreased the relative oral bioavailability of DON (39.9%), OTA (44.3%), and AFB1 (64.1%). In experiment 2, one-day-old male Ross broilers (n = 600) were divided into three treatments with ten replicates. Each replicate was a pen with 20 birds. The broiler chickens were fed a control diet with negligible levels of DON (0.19-0.25 mg/kg) or diets naturally contaminated with moderate levels of DON (2.60-2.91 mg/kg), either supplemented or not with an algoclay-based decontaminant (2 g/kg diet). Jejunum villus damage was observed on day 28, followed by villus shortening on d37 in broiler chickens fed the DON-contaminated diet. This negative effect was not observed when the DON-contaminated diet was supplemented with the algoclay-based decontaminant. On d37, the mRNA expression of glutathione synthetase was significantly increased in the liver of broiler chickens fed the DON-contaminated diet. However, its expression was similar to the control when the birds were fed the DON-contaminated diet supplemented with the algoclay-based decontaminant. In conclusion, the algoclay-based decontaminant reduced the systemic exposure of broiler chickens to DON, OTA, and AFB1 in a single oral bolus model. This can be attributed to the binding of the mycotoxins in the gastrointestinal tract. Moreover, dietary contamination with DON at levels between 2.69 and 2.91 mg/kg did not impair production performance but had a negative impact on broiler chicken intestinal morphology and the liver redox system. When the algoclay-based decontaminant was added to the diet, the harm caused by DON was no longer observed. This correlates with the results obtained in the toxicokinetic assay and can be attributed to a decreased absorption of DON.


Assuntos
Aflatoxina B1 , Ração Animal , Galinhas , Contaminação de Alimentos , Fígado , Ocratoxinas , Estresse Oxidativo , Tricotecenos , Animais , Tricotecenos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Masculino , Ocratoxinas/toxicidade , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Aflatoxina B1/toxicidade , Ração Animal/análise , Intestinos/efeitos dos fármacos , Intestinos/patologia , Toxicocinética , Dieta/veterinária , Silicatos de Alumínio
16.
Sci Total Environ ; 935: 173285, 2024 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-38772488

RESUMO

Dietary pollution of Aflatoxin B1 (AFB1) poses a great threat to global food safety, which can result in serious hepatic injuries. Following the widespread use of plastic tableware, co-exposure to microplastics and AFB1 has dramatically increased. However, whether microplastics could exert synergistic effects with AFB1 and amplify its hepatotoxicity, and the underlying mechanisms are still unelucidated. Here, mice were orally exposed to 100 nm polystyrene nanoplastics (NPs) and AFB1 to investigate the influences of NPs on AFB1-induced hepatic injuries. We found that exposure to only NPs or AFB1 resulted in colonic inflammation and the impairment of the intestinal barrier, which was exacerbated by combined exposure to NPs and AFB1. Meanwhile, co-exposure to NPs exacerbated AFB1-induced dysbiosis of gut microbiota and remodeling of the fecal metabolome. Moreover, NPs and AFB1 co-exposure exhibited higher levels of systemic inflammatory factors compared to AFB1 exposure. Additionally, NPs co-exposure further exacerbated AFB1-induced hepatic fibrosis and inflammation, which could be associated with the overactivation of the TLR4/MyD88/NF-κB pathway. Notably, Spearman's correlation analysis revealed that the exacerbation of NPs co-exposure was closely associated with microbial dysbiosis. Furthermore, microbiota from NPs-exposed mice (NPsFMT) partly reproduced the exacerbation of NPs on AFB1-induced systemic and hepatic inflammation, but not fibrosis. In summary, our findings indicate that gut microbiota could be involved in the exacerbation of NPs on AFB1-induced hepatic injuries, highlighting the health risks of NPs.


Assuntos
Aflatoxina B1 , Microbioma Gastrointestinal , Fígado , Microplásticos , Poliestirenos , Aflatoxina B1/toxicidade , Animais , Camundongos , Microbioma Gastrointestinal/efeitos dos fármacos , Poliestirenos/toxicidade , Microplásticos/toxicidade , Fígado/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas , Disbiose/induzido quimicamente , Nanopartículas/toxicidade
17.
Toxicon ; 244: 107770, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38768829

RESUMO

Aflatoxins are toxic compounds produced by certain molds, primarily Aspergillus species, which can contaminate crops such as grains and nuts. These toxins pose a significant health risk to animals and humans. Aflatoxin B1 (AFB1) is the most potent of these compounds and has been well-characterized to lead to diminished growth and feed efficiency by disrupting nutrient absorption and metabolism in poultry. AFB1 can trigger apoptosis and inflammation, leading to a decline in immune function and changes in blood biochemistry in poultry. Recently, there has been growing interest in using microalgae as a natural antioxidant to mitigate the effects of aflatoxins in poultry diets. Microalgae have strong antioxidant, antimicrobial, anti-apoptotic, and anti-inflammatory properties, and adding them to aflatoxin-contaminated poultry diets has been shown to improve growth and overall health. This review investigates the potential of microalgae, such as Spirulina platensis, Chlorella vulgaris, and Enteromorpha prolifera, to mitigate AFB1 contamination in poultry feeds. These microalgae contain substantial amounts of bioactive compounds, including polysaccharides, peptides, vitamins, and pigments, which possess antioxidant, antimicrobial, and detoxifying properties. Microalgae can bind to aflatoxins and prevent their absorption in the gastrointestinal tract of poultry. They can also enhance the immune system of poultry, making them more resilient to the toxic effects of AFB1. Based on the data collected, microalgae have shown promising results in combating AFB1 contamination in poultry feeds. They can bind to aflatoxins, boost the immune system, and improve feed quality. This review emphasizes the harmful effects of AFB1 on poultry and the promising role of microalgae in reducing these effects.


Assuntos
Aflatoxina B1 , Ração Animal , Microalgas , Aves Domésticas , Animais , Aflatoxina B1/toxicidade , Contaminação de Alimentos/prevenção & controle , Antioxidantes/farmacologia , Spirulina , Aflatoxinas/toxicidade
18.
Int J Mol Sci ; 25(10)2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38791343

RESUMO

AIMS: The current review aims to outline and summarize the latest research on aflatoxin, with research studies describing natural, herbal and chemical compound applications in animal (pig) models and in vitro cellular studies. Aflatoxin, a carcinogenic toxin metabolite, is produced by Aspergillus flavus in humid environments, posing a threat to human health and crop production. The current treatment involves the prevention of exposure to aflatoxin and counteracting its harmful toxic effects, enabling survival and research studies on an antidote for aflatoxin. OBJECTIVES: To summarize current research prospects and to outline the influence of aflatoxin on animal forage in farm production, food and crop processing. The research application of remedies to treat aflatoxin is undergoing development to pinpoint biochemical pathways responsible for aflatoxin effects transmission and actions of treatment. SIGNIFICANCE: To underline the environmental stress of aflatoxin on meat and dairy products; to describe clinical syndromes associated with aflatoxicosis on human health that are counteracted with proposed treatment and preventive interventions. To understand how to improve the health of farm animals with feed conditions.


Assuntos
Aflatoxina B1 , Ração Animal , Contaminação de Alimentos , Animais , Humanos , Aflatoxina B1/toxicidade , Aflatoxina B1/efeitos adversos , Contaminação de Alimentos/prevenção & controle , Aspergillus flavus/metabolismo , Aspergillus flavus/efeitos dos fármacos
19.
Ecotoxicol Environ Saf ; 278: 116336, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38691883

RESUMO

Aflatoxin B1 (AFB1), a common mycotoxin, can occur in agricultural products. As a metabolite of AFB1, aflatoxin M1 (AFM1) mainly exist in dairy products. These two mycotoxins threaten human health, although it is unclear how they affect the function of the intestinal barrier. In this study, mice were exposed to AFB1 (0.3 mg/kg body b.w.) and AFM1(3.0 mg/kg b.w.) either individually or in combination for 28 days to explore the main differentially expressed proteins (DEPs) and the associated enriched pathways. These findings were preliminarily verified by the transcriptomic and proteomic analyses in differentiated Caco-2 cells. The results revealed that AFB1 and AFM1 exposure in mice disrupted the function of the intestinal barrier, and the combined toxicity was greater than that of each toxin alone. Further proteomic analysis in mice demonstrated that the mechanisms underlying these differences could be explained as follows: (i) lipid metabolism was enriched by AFB1-induced DEPs. (ii) protein export pathway was stimulated by AFM1-induced DEPs. (iii) cell metabolic ability was inhibited (as evidenced by changes in UDP-GT1, UDP-GT2, and Gatm6), apoptosis was induced (MAP4K3), and epithelial cell integrity was disrupted (Claudin7 and IQGAP2), resulting in more extensive intestinal damage after combined treatment. In conclusion, the hazardous impact of co-exposure to AFB1 and AFM1 from proteomic perspectives was demonstrated in the present study.


Assuntos
Aflatoxina B1 , Aflatoxina M1 , Proteômica , Aflatoxina M1/toxicidade , Aflatoxina B1/toxicidade , Animais , Camundongos , Células CACO-2 , Humanos , Masculino , Intestinos/efeitos dos fármacos , Intestinos/patologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo
20.
Food Chem Toxicol ; 188: 114640, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38583501

RESUMO

This study investigates the individual and combined effects of the mycotoxins, Aflatoxin B1 (AFB1), Enniatin B (ENNB) and Sterigmatocystin (STG), on the cellular viability of gastric (NCI-N87), intestinal (Caco-2), hepatic (Hep-G2) and renal (Hek-293) cells, shedding light on synergistic or antagonistic effects using a constant ratio combination design proposed by Chou-Talalay. These toxins are prevalent in cereal-based foods, frequently consumed by children which raises concerns about their exposure to these mycotoxins. This population is particularly vulnerable to the effects of these toxins due to their underdeveloped organs and incompletely structured physiological processes. Results showed that ENB was the most toxic of the three mycotoxins across all cell lines, while STG and AFB1 showed lower toxicity. The combination of ENNB + STG was found to be the most potent in terms of binary mixtures. In regard to ternary combinations, Caco-2 cells are more sensitive to the tested mycotoxins, whereas NCI-N87 cells show lower levels of cell damage. Worrying dose reduction values (>10-fold) were found for ENNB in binary and ternary combinations at low exposure levels. These findings are significant for establishing initial reference values, which play a pivotal role in estimating reference doses that are subsequently incorporated into the broader risk assessment process.


Assuntos
Aflatoxina B1 , Depsipeptídeos , Esterigmatocistina , Humanos , Esterigmatocistina/toxicidade , Aflatoxina B1/toxicidade , Depsipeptídeos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Células CACO-2 , Fígado/efeitos dos fármacos , Rim/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Células HEK293 , Células Hep G2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...