Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 455
Filtrar
1.
Mutagenesis ; 39(3): 181-195, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38468450

RESUMO

Aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are mycotoxins widely distributed in maize and maized-based products, often occurring together. The implications of co-exposure to aflatoxin and fumonsin for human health are numerous, but a particular concern is the potential of FB1 to modulate AFB1 hepatotoxicity. This study evaluated the toxicity of these mycotoxins, alone or combined, in a human non-tumorigenic liver cell line, HHL-16 cells, and assessed the effects of AFB1 and FB1 on expression of genes involved in immune and growth factor pathways. The results demonstrated that in HHL-16 cells, both AFB1 and FB1 had dose-dependent and time-dependent toxicity, and the combination of them showed a synergistic toxicity in the cells. Moreover, AFB1 caused upregulation of IL6, CCL20, and BMP2, and downregulation of NDP. In combination of AFB1 with FB1, gene expression levels of IL6 and BMP2 were significantly higher compared to individual FB1 treatment, and had a tendency to be higher than individual AFB1 treatment. This study shows that FB1 may increase the hepatoxicity of AFB1 through increasing the inflammatory response and disrupting cell growth pathways.


Assuntos
Aflatoxina B1 , Fumonisinas , Hepatócitos , Fumonisinas/toxicidade , Humanos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Aflatoxina B1/toxicidade , Linhagem Celular , Inflamação/genética , Inflamação/induzido quimicamente , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo
2.
J Food Sci ; 89(2): 1280-1293, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38193205

RESUMO

The Fusarium verticillioides produces a mycotoxin, that is, fumonisin b1 (Fb1), which commonly infects corn and agricultural commodities. The Fb1 showed hepatotoxicity, neurotoxicity, and carcinogenicity in animals. Hence, the present investigation aimed to evaluate the effect of apocynin (AP) on Fb1-induced neurotoxic effects and its mechanism in the mice model and cell line. The male Balb/c mice, with the 6.75 mg/kg bwt of Fb1 were injected subcutaneously for 5 days to induce neurotoxicity. A significant elevation of serotonin (5-HT) was observed in mice treated with Fb1 in the whole brain showing biogenic amines may reflect Fb1 neurotoxicity, but the negatively regulated mechanisms were attenuated by the pretreatment of AP. In addition, AP pretreatment normalized apoptotic changes in histology and immunohistochemistry studies. In Western blotting studies, apoptotic genes were upregulated and oxidative stress genes were downregulated due to Fb1 treatment; while treating with AP, these gene expressions were rectified. Further cell cytotoxicity was investigated by MTT and lactate dehydrogenase (LDH) assays in SH-SY5Y cell line. MTT and LDH assays indicated the IC50 value to be 150 µM of Fb1, which was protected by 100 µg of AP. The electron microscopy evaluated the Fb1-induced apoptotic conditions and its cell morphology recovery by AP. These results suggest that nicotinamide adenine dinucleotide phosphate hydrogen oxidase-mediated reactive oxygen species is the primary upstream signal leading to increased Fb1-mediated neurotoxicity in mice. The use of the antioxidant AP reversed the toxin-induced oxidative stress and apoptosis by its antioxidant potency.


Assuntos
Acetofenonas , Fumonisinas , Neuroblastoma , Fármacos Neuroprotetores , Humanos , Masculino , Camundongos , Animais , Fármacos Neuroprotetores/farmacologia , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Fumonisinas/toxicidade , Fumonisinas/metabolismo , Apoptose , Estresse Oxidativo , Modelos Animais
3.
Environ Toxicol ; 39(2): 905-914, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37955343

RESUMO

Fumonisins are common contaminants in the global food and environment, pose a variety of health risks to humans and animals. However, the method of mitigating fumonisin toxicity is still unclear. Resveratrol is a natural compound with antioxidant and anti-inflammatory properties. In this study, the protective effect of resveratrol against fumonisin-induced intestinal toxicity was investigated by the porcine intestinal epithelial cell line (IPEC-J2). The cells were treated with 0-40 µM fumonisin for 24 or 48 h with or without the 24 h resveratrol (15 µM) pretreatment. The data showed that resveratrol could alleviate the fumonisin B1 (FB1)-induced decrease in cell viability and amplify in membrane permeability. At the same time, it could reduce the accumulation of intracellular reactive oxygen species and increase the expression ranges of Nrf2 and downstream genes (SOD1 and NQO-1), thereby counteracting FB1-induced apoptosis. Furthermore, resveratrol was able to reduce the expression levels of inflammatory factors (TNF-α, IL-1ß, and IL-6), increase the expression levels of tight junction proteins (Claudin-1, Occludin, and ZO-1), and the integrity of the IPEC-J2 monolayer. Our data also showed that resveratrol could attenuate the toxicity of the co-occurrence of three fumonisins. It is implied that resveratrol represents a promising protective approach for fumonisin, even other mycotoxins in the future. This provided a new strategy for further blocking and controlling the toxicity of fumonisin, subsequently avoiding adverse effects on the human and animal health.


Assuntos
Fumonisinas , Animais , Suínos , Humanos , Fumonisinas/toxicidade , Fumonisinas/metabolismo , Resveratrol/farmacologia , Junções Íntimas/metabolismo , Células Epiteliais , Inflamação/induzido quimicamente , Inflamação/metabolismo , Apoptose
4.
Appl Environ Microbiol ; 89(12): e0121123, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-38054733

RESUMO

IMPORTANCE: Fumonisins can cause diseases in animals and humans consuming Fusarium-contaminated food or feed. The search for microbes capable of fumonisin degradation, or for enzymes that can detoxify fumonisins, currently relies primarily on chemical detection methods. Our constructed fumonisin B1-sensitive yeast strain can be used to phenotypically detect detoxification activity and should be useful in screening for novel fumonisin resistance genes and to elucidate fumonisin metabolism and resistance mechanisms in fungi and plants, and thereby, in the long term, help to mitigate the threat of fumonisins in feed and food.


Assuntos
Fumonisinas , Fusarium , Humanos , Animais , Fumonisinas/toxicidade , Fumonisinas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ração Animal , Fusarium/genética , Fusarium/metabolismo
5.
Nutrients ; 15(21)2023 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-37960333

RESUMO

In this study, the soluble, but non-digestible, longan (Dimocarpus longan Lour.) polysaccharides (LP) were extracted from dried longan fruits and then chemically selenylated to produce two selenylated products, namely SeLP1 and SeLP2, with different selenylation extents. The aim was to investigate their protective effects on rat intestinal epithelial (IEC-6) cells exposed to the food toxin fumonisin B1 (FB1). LP only contained total Se content of less than 0.01 g/kg, while SeLP1 and SeLP2 were measured with respective total Se content of up to 1.46 and 4.79 g/kg. The cell viability results showed that these two selenylated products were more efficient than LP in the IEC-6 cells in alleviating FB1-induced cell toxicity, suppressing lactate dehydrogenase (LDH) release, and decreasing the generation of intracellular reactive oxygen species (ROS). These two selenylated products were also more effective than LP in combating FB1-induced barrier disruption via increasing the transepithelial electric resistance (TEER), reducing the paracellular permeability, decreasing the mitochondrial membrane potential (MMP) loss, and maintaining cell barrier integrity by upregulating the tight-junction-related genes and proteins. FB1 caused cell oxidative stress and barrier dysfunction by activating the MAPK and mitochondrial apoptosis signaling pathways, while SeLP1 and SeLP2 could regulate the tMAPK- and apoptosis-related proteins to suppress the FB1-mediated activation of the two pathways. Overall, SeLP2 was observed to be more active than SeLP1 in the IEC-6 cells. In conclusion, the chemical selenylation of LP caused an activity enhancement to ameliorate the FB1-induced cell cytotoxicity and intestinal barrier disruption. Meanwhile, the increased selenylation of LP would endow the selenylated product SeLP2 with more activity.


Assuntos
Fumonisinas , Sapindaceae , Ratos , Animais , Fumonisinas/farmacologia , Fumonisinas/toxicidade , Intestinos , Células Epiteliais
6.
Ecotoxicol Environ Saf ; 268: 115697, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37979349

RESUMO

The biological properties of sphinganine-(d18:0)-, sphingosine-(d18:1)-, deoxysphinganine-(m18: 0)-, deoxysphingosine-(m18:1)-, deoxymethylsphinganine-(m17:0)-, deoxymethylsphingosine-(m17:1)-, sphingadienine-(d18:2)-, and phytosphingosine-(t18:0)-sphingolipids have been reported to vary, but little is known about the effects of fumonisins, which are mycotoxins that inhibit ceramide synthase, on sphingolipids other than those containing d18:0 and d18:1. Thirty chickens divided into three groups received a control diet or a diet containing 14.6 mg FB1 + FB2/kg for 14 and 21 days. No effects on health or performance were observed, while the effects on sphingoid bases, ceramides, sphingomyelins, and glycosylceramides in liver, kidney, and plasma varied. The t18:0 forms were generally unaffected by fumonisins, while numerous effects were found for m18:0, m18:1, d18:2, and the corresponding ceramides, and these effects appeared to be similar to those observed for d18:0-, and d18:1-ceramides. Partial least square discriminant analysis showed that d18:1- and d18:0-sphingolipids are important variables for explaining the partitioning of chickens into different groups according to fumonisins feeding, while m17:1-, m18:0-, m18:1-, d18:2-, and t18:0-sphingolipids are not. Interestingly, the C22-C24:C16 ratios measured for each class of sphingolipid increased in fumonisin-fed chickens in the three assayed matrices, whereas the total amounts of the sphingolipid classes varied. The potential use of C22-C24:C16 ratios as biomarkers requires further study.


Assuntos
Fumonisinas , Animais , Fumonisinas/toxicidade , Galinhas , Esfingolipídeos , Ceramidas , Fígado , Rim
7.
Toxins (Basel) ; 15(11)2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37999492

RESUMO

Mycotoxins present a significant health concern within the animal-feed industry, with profound implications for the pig-farming sector. The objective of this study was to evaluate the efficacy of two commercial adsorbents, an organically modified clinoptilolite (OMC) and a multicomponent mycotoxin detoxifying agent (MMDA), to ameliorate the combined adverse effects of dietary aflatoxins (AFs: sum of AFB1, AFB2, AFG1, and AFG2), fumonisins (FBs), and zearalenone (ZEN) at levels of nearly 0.5, 1.0, and 1.0 mg/kg, on a cohort of cross-bred female pigs (N = 24). Pigs were randomly allocated into six experimental groups (control, mycotoxins (MTX) alone, MTX + OMC 1.5 kg/ton, MTX + OMC 3.0 kg/ton, MTX + MMDA 1.5 kg/ton, and MTX + MMDA 3.0 kg/ton), each consisting of four individuals, and subjected to a dietary regimen spanning 42 days. The administration of combined AFs, FBs, and ZEN reduced the body-weight gain and increased the relative weight of the liver, while there was no negative influence observed on the serum biochemistry of animals. The supplementation of OMC and MMDA ameliorated the toxic effects, as observed in organ histology, and provided a notable reduction in residual AFs, FBs, and ZEN levels in the liver and kidneys. Moreover, the OMC supplementation was able to reduce the initiation of liver carcinogenesis without any hepatotoxic side effects. These findings demonstrate that the use of OMC and MMDA effectively mitigated the adverse effects of dietary AFs, FBs, and ZEN in piglets. Further studies should explore the long-term protective effects of the studied adsorbent supplementation to optimize mycotoxin management strategies in pig-farming operations.


Assuntos
Aflatoxinas , Fumonisinas , Micotoxinas , Zearalenona , Animais , Feminino , Humanos , Aflatoxinas/toxicidade , Contaminação de Alimentos/prevenção & controle , Contaminação de Alimentos/análise , Fumonisinas/toxicidade , Micotoxinas/análise , Suínos , Zearalenona/análise
8.
Toxins (Basel) ; 15(11)2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37999516

RESUMO

Mycotoxins are considered the most threating natural contaminants in food. Among these mycotoxins, aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are the most prominent fungal metabolites that represent high food safety risks, due to their widespread co-occurrence in several food commodities, and their profound toxic effects on humans. Considering the ethical and more humane animal research, the 3Rs (replacement, reduction, and refinement) principle has been promoted in the last few years. Therefore, this review aims to summarize the research studies conducted up to date on the toxicological effects that AFB1 and FB1 can induce on human health, through the examination of a selected number of in vitro studies. Although the impact of both toxins, as well as their combination, were investigated in different cell lines, the majority of the work was carried out in hepatic cell lines, especially HepG2, owing to the contaminants' liver toxicity. In all the reviewed studies, AFB1 and FB1 could invoke, after short-term exposure, cell apoptosis, by inducing several pathways (oxidative stress, the mitochondrial pathway, ER stress, the Fas/FasL signaling pathway, and the TNF-α signal pathway). Among these pathways, mitochondria are the primary target of both toxins. The interaction of AFB1 and FB1, whether additive, synergistic, or antagonistic, depends to great extent on FB1/AFB1 ratio. However, it is generally manifested synergistically, via the induction of oxidative stress and mitochondria dysfunction, through the expression of the Bcl-2 family and p53 proteins. Therefore, AFB1 and FB1 mixture may enhance more in vitro toxic effects, and carry a higher significant risk factor, than the individual presence of each toxin.


Assuntos
Fumonisinas , Micotoxinas , Animais , Humanos , Aflatoxina B1/toxicidade , Fumonisinas/toxicidade , Micotoxinas/toxicidade , Fígado
9.
Toxins (Basel) ; 15(11)2023 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-37999527

RESUMO

Fumonisin B1 (FB1), a mycotoxin produced by Fusarium verticillioides, is one of the most common pollutants in natural foods and agricultural crops. It can cause chronic and severe health issues in humans and animals. The aim of this study was to evaluate the transgenerational effects of FB1 exposure on the structure and function of the kidneys in offspring. Virgin female Wistar rats were randomly divided into three groups: group one (control) received sterile water, and groups two and three were intragastrically administered low (20 mg/kg) and high (50 mg/kg) doses of FB1, respectively, from day 6 of pregnancy until delivery. Our results showed that exposure to either dose of FB1 caused histopathological changes, such as atrophy, hypercellularity, hemorrhage, calcification, and a decrease in the glomerular diameter, in both the first and second generations. The levels of the antioxidant markers glutathione, glutathione S-transferase, and catalase significantly decreased, while malondialdehyde levels increased. Moreover, autophagy was induced, as immunofluorescence analysis revealed that LC-3 protein expression was significantly increased in both generations after exposure to either dose of FB1. However, a significant decrease in methyltransferase (DNMT3) protein expression was observed in the first generation in both treatment groups (20 mg/kg and 50 mg/kg), indicating a decrease in DNA methylation as a result of early-life exposure to FB1. Interestingly, global hypomethylation was also observed in the second generation in both treatment groups despite the fact that the mothers of these rats were not exposed to FB1. Thus, early-life exposure to FB1 induced nephrotoxicity in offspring of the first and second generations. The mechanisms of action underlying this transgenerational effect may include oxidative stress, autophagy, and DNA hypomethylation.


Assuntos
Fumonisinas , Micotoxinas , Humanos , Ratos , Feminino , Animais , Micotoxinas/toxicidade , Metilação de DNA , Ratos Wistar , Fumonisinas/toxicidade , Estresse Oxidativo , Autofagia , DNA
10.
Environ Sci Pollut Res Int ; 30(53): 114438-114451, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37858030

RESUMO

Fumonisin B1 (FB1) is a widely present mycotoxin that accumulates in biological systems and poses a health risk to animals. However, few studies have reported the molecular mechanism by which FB1 induces nephrotoxicity. The aim of this study was to assess the extent of nephrotoxicity during FB1 exposure and the possible molecular mechanisms behind it. Therefore, 180 young quails were equally divided into two groups. The control group was fed typical quail food, while the experimental group was fed quail food containing 30 mg·kg-1 FB1. Various parameters were assessed, which included histopathological, ultrastructural changes, levels of biochemical parameters, oxidative indicators, inflammatory factors, possible target organelles mitochondrial and endoplasmic reticulum (ER)-related factors, nuclear xenobiotic receptors (NXR) response, and cytochrome P450 system (CYP450s)-related factors in the kidneys on days 14, 28, and 42. The results showed that FB1 can induce oxidative stress through NXR response and disorder of the CYP450s system, leading to mitochondrial dysfunction and ER stress, promoting the expression of inflammatory factors (including IL-1ß, IL-6, and IL-8) and causing kidney damage. This study elucidated the possible molecular mechanism by which FB1 induces nephrotoxicity in young quails.


Assuntos
Fumonisinas , Micotoxinas , Animais , Codorniz , Fígado/metabolismo , Fumonisinas/toxicidade , Micotoxinas/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo
11.
J Agric Food Chem ; 71(36): 13462-13473, 2023 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-37655855

RESUMO

The current study evaluated the effects of feeding diets contaminated with aflatoxin B1 (AFB1), fumonisins (FBs), or both on the performance and health of broiler chickens and the safety of their food products as well as the efficacy of bentonite and fumonisin esterase to mitigate the effects of these mycotoxins under conditions representative for sub-Saharan Africa (SSA). Four hundred one-day-old Cobb 500 broiler chickens were randomly assigned to 20 treatments with either a control diet, a diet with moderate AFB1 (60 µg/kg feed) or high AFB1 (220 µg/kg feed), or FBs (17,430 µg FB1+FB2/kg feed), alone or in combination, a diet containing AFB1 (either 60 or 220 µg/kg) and/or FBs (17,430 µg FB1+FB2/kg) and bentonite or fumonisin esterase or both, or a diet with bentonite or fumonisin esterase only. The experimental diets were given to the birds from day 1 to day 35 of age, and the effects of the different treatments on production performance were assessed by feed intake (FI), body weight gain (BWG), and feed conversion ratio (FCR). Possible health effects were evaluated through blood biochemistry, organ weights, mortality, liver gross pathological changes, and vaccine response. Residues of aflatoxins (AFB1, B2, G1, G2, M1 and M2) were determined in plasma, muscle, and liver tissues using validated UHPLC-MS/MS methods. The results obtained indicated that broiler chickens fed high AFB1 alone had poor FCR when compared to a diet with both high AFB1 and FBs (p = 0.0063). Serum total protein and albumin from birds fed FBs only or in combination with moderate or high AFB1 or detoxifiers increased when compared to the control (p < 0.05). Liver gross pathological changes were more pronounced in birds fed contaminated diets when compared to birds fed the control or diets supplemented with mycotoxin detoxifiers. The relative weight of the heart was significantly higher in birds fed high AFB1 and FBs when compared to the control or high AFB1 only diets (p < 0.05), indicating interactions between the mycotoxins. Inclusion of bentonite in AFB1-contaminated diets offered a protective effect on the change in weights of the liver, heart and spleen (p < 0.05). Residues of AFB1 were detected above the limit of quantification (max: 0.12 ± 0.03 µg/kg) in liver samples only, from birds fed a diet with high AFB1 only or with FBs or the detoxifiers. Supplementing bentonite into these AFB1-contaminated diets reduced the levels of the liver AFB1 residues by up to 50%. Bentonite or fumonisin esterase, alone, did not affect the performance and health of broiler chickens. Thus, at the doses tested, both detoxifiers were safe and efficient for use as valid means of counteracting the negative effects of AFB1 and FBs as well as transfer of AFB1 to food products (liver) of broiler chickens.


Assuntos
Aflatoxinas , Fumonisinas , Micotoxinas , Animais , Aflatoxinas/toxicidade , Galinhas , Fumonisinas/toxicidade , Bentonita , Espectrometria de Massas em Tandem , Aflatoxina B1/toxicidade , Esterases
12.
Toxicol In Vitro ; 93: 105670, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37633472

RESUMO

The presence of mycotoxins such as Fumonisin B1(FB1) and Ochratoxin A (OTA) in food and feed has become a threat to human and animal health since they can produce several afflictions. Different mechanisms of action by which they exercise their cytotoxic activity have been attributed to them, including the production of reactive oxygen species (ROS). For this reason, a measurement of the production of ROS species, and an evaluation of the intrinsic cell enzymatic antioxidant activity, including glutathione peroxidase (GPx), glutathione transferase (GTS), and catalase (CAT) together with a cytotoxicity and cell cycle assay have been performed in undifferentiated SH-SY5Y cells exposed to FB1, OTA and [FB1 + OTA] after 24 h and 48 h. FB1 and OTA. Monitoring of intracellular ROS production was carried out by the H2-DCFDA probe; while spectrometry analysis of absorbances was used for measuring GPx, GST and CAT activity. Finally, cell proliferation and cell cycle distribution were studied by flow cytometry. When cells were treated with OTA, an increase in GPx and GST activity was observed compared to FB1 and [FB1 + OTA]; conversely, a decrease in CAT activity was observed when cells were exposed to OTA coinciding with the results observed for ROS measurement. Regarding the cell cycle, when cells were exposed to OTA, a decrease in G0/G1 was detected, revealing an arrest of cell division for SH-SY5Y cells at the concentrations studied.


Assuntos
Fumonisinas , Micotoxinas , Neuroblastoma , Animais , Humanos , Espécies Reativas de Oxigênio/metabolismo , Fumonisinas/toxicidade , Micotoxinas/toxicidade , Antioxidantes , Divisão Celular , Ciclo Celular
13.
Arch Toxicol ; 97(10): 2707-2719, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37589943

RESUMO

Contamination with fumonisins produced by Fusarium spp. is rapidly growing in both developing and developed countries. The purpose of this study was to determine whether oral exposure to fumonisin contributed to the development of allergic diseases. We initially examined the immunotoxic potential of short-term, oral administration of fumonisin B1 (FB1, 1 mg/kg) and fumonisin B2 (FB2, 1 mg/kg), both naturally occurring fumonisins, using a BALB/c mouse model of allergic contact dermatitis and Dermatophagoides farina-induced asthma. Using an NC/nga mouse model of atopic dermatitis (AD), we evaluated the adverse effects of subchronic oral exposure to low concentrations of FB2 (2 or 200 µg/kg). Finally, we explored the influence of FB2 on regulatory T cell proliferation and function in mesenteric lymph nodes after 1-week oral exposure to FB2 in BALB/c mice. Oral exposure to FB2 markedly exacerbated the symptoms of allergy, including skin thickness, histological evaluation, immunocyte proliferation, and proinflammatory cytokine production, although no change was observed following exposure to FB1. Furthermore, oral exposure to low concentrations of FB2 considerably exacerbated the AD scores, skin thickness, transepidermal water loss, histological features, and proinflammatory cytokine production. The aggravated allergic symptoms induced by oral exposure to FB2 could be attributed to the direct inhibition of IL-10 production by regulatory T cells in mesenteric lymph nodes. Our findings indicate that the recommended maximum fumonisin level should be reconsidered based on the potential for allergy development.


Assuntos
Dermatite Alérgica de Contato , Fumonisinas , Animais , Camundongos , Fumonisinas/toxicidade , Interleucina-10 , Linfócitos T Reguladores , Linfonodos
14.
Toxins (Basel) ; 15(7)2023 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-37505702

RESUMO

This study aimed to investigate the efficacy of a feed additive containing bentonite and enzymatically hydrolyzed yeast on the intestinal health and growth of newly weaned pigs under chronic dietary exposure to fumonisin and aflatoxin. Newly weaned pigs were randomly allotted to one of four possible treatments: a control diet of conventional corn; a diet of corn contaminated with fumonisin and aflatoxin; a diet of mycotoxin-contaminated corn with 0.2% of feed additive; and a diet of mycotoxin contaminated corn with 0.4% of feed additive. We observed lower average weight gain and average daily feed intake in pigs that were fed only mycotoxin-contaminated corn compared to the control group. Feed additive supplementation linearly increased both average weight gain and feed intake, as well as tumor necrosis factor-alpha. In the jejunum, there was an observed decrease in immunoglobulin A and an increase in claudin-1. Additionally, feed additive supplementation increased the villus height to crypt depth ratio compared to the control. In conclusion, feed additives containing bentonite and enzymatically hydrolyzed yeast could mitigate the detrimental effects of mycotoxins on the growth performance of newly weaned pigs by improving intestinal integrity and positively modulating immune response.


Assuntos
Aflatoxinas , Fumonisinas , Micotoxinas , Suínos , Animais , Fumonisinas/toxicidade , Saccharomyces cerevisiae , Bentonita , Aflatoxinas/toxicidade , Suplementos Nutricionais/análise , Dieta/veterinária , Micotoxinas/toxicidade , Aumento de Peso , Ração Animal/análise
15.
Mycotoxin Res ; 39(3): 271-283, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37328702

RESUMO

Fumonisin B1 (FB1) poses a risk to animal and human health. Although the effects of FB1 on sphingolipid metabolism are well documented, there are limited studies covering the epigenetic modifications and early molecular alterations associated with carcinogenesis pathways caused by FB1 nephrotoxicity. The present study investigates the effects of FB1 on global DNA methylation, chromatin-modifying enzymes, and histone modification levels of the p16 gene in human kidney cells (HK-2) after 24 h exposure. An increase (2.23-fold) in the levels of 5-methylcytosine (5-mC) at 100 µmol/L was observed, a change independent from the decrease in gene expression levels of DNA methyltransferase 1 (DNMT1) at 50 and 100 µmol/L; however, DNMT3a and DNMT3b were significantly upregulated at 100 µmol/L of FB1. Dose-dependent downregulation of chromatin-modifying genes was observed after FB1 exposure. In addition, chromatin immunoprecipitation results showed that 10 µmol/L of FB1 induced a significant decrease in H3K9ac, H3K9me3 and H3K27me3 modifications of p16, while 100 µmol/L of FB1 caused a significant increase in H3K27me3 levels of p16. Taken together, the results suggest that epigenetic mechanisms might play a role in FB1 carcinogenesis through DNA methylation, and histone and chromatin modifications.


Assuntos
Cromatina , Fumonisinas , Humanos , Fumonisinas/toxicidade , Genes p16 , Código das Histonas , Histonas , Rim/metabolismo
16.
Arh Hig Rada Toksikol ; 74(2): 115-119, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37357877

RESUMO

The aim of this study was to investigate the effects of resveratrol against fumonisin B1 (FB1)-induced liver toxicity, as, to the best of our knowledge, these effects have not been investigated yet, even though the toxic effects and mechanisms of FB1 and the antioxidative effects of resveratrol are well known. 40 BALB/c mice were divided into control, FB1, resveratrol, and FB1+resveratrol groups. Control received saline for 14 days. The FB1 group received 2.25 mg/kg FB1 every other day for 14 days. The resveratrol group received 10 mg/kg resveratrol for 14 days. The FB1+resveratrol group received 2.25 mg/kg FB1 every other day and 10 mg/kg resveratrol every day for 14 days. All administrations were peritoneal. Alanine aminotransferase (ALT), aspartate aminotransferase (AST), total sialic acid (TSA) levels were analysed in serum samples, while total antioxidant status (TAS) and total oxidant status (TOS) were measured in the liver. Additionally, the liver tissue was examined for histopathological changes. AST, ALT, and TSA were significantly higher in the FB1 group than control. Resveratrol countered FB1 effects for all parameters, including TOS and TAS. Liver histology showed FB1-induced hyperaemia, infiltrations, and megalokaryosis in some hepatocytes. No pathological findings were detected in the control, resveratrol, or FB1+resveratrol group. Our findings confirm resveratrol's protective effect against liver damage and oxidative stress caused by FB1. In addition, they suggest that increased serum TSA levels can be used as a biomarker of FB1-induced hepatotoxicity.


Assuntos
Fumonisinas , Camundongos , Animais , Resveratrol/farmacologia , Resveratrol/metabolismo , Fumonisinas/toxicidade , Fumonisinas/metabolismo , Fígado , Estresse Oxidativo
17.
Environ Int ; 175: 107945, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37126917

RESUMO

Aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are widely (co-)detected in food and known for their hepatotoxicity in humans. Still, their combined toxicity needs to be investigated, especially the impact on mitochondria. In our previous work, we examined the effect of short-term exposure to different doses of AFB1, FB1, and their binary mixture (MIX) on the bioenergetic status of HepG2 cells, a well-recognized in vitro model system for studying liver cell function. In the current work, we further investigated the (combined) effect of AFB1 and FB1 on the mitochondrial and glycolytic activity of HepG2 cells using Seahorse respirometry analysis and RNA transcriptome sequencing. The results showed that the co-exposure, especially at high doses, is more toxic due to a more inhibition of all parameters of mitochondrial respiration. However, FB1 contributes more to the MIX effects than AFB1. RNA transcriptome sequencing showed that the p53 signaling pathway, a major orchestrator of mitochondrial apoptosis, was differentially expressed. Moreover, the co-exposure significantly downregulated the genes encoding for Complexes I, II, III, and IV, representing the onset of the suppressed mitochondrial respiration in HepG2 cells.


Assuntos
Aflatoxina B1 , Fumonisinas , Humanos , Aflatoxina B1/toxicidade , Células Hep G2 , Transcriptoma , Fumonisinas/toxicidade
18.
Biomolecules ; 13(5)2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37238625

RESUMO

Fumonisins are frequent food contaminants. The high exposure to fumonisins can cause harmful effects in humans and animals. Fumonisin B1 (FB1) is the most typical member of this group; however, the occurrence of several other derivatives has been reported. Acylated metabolites of FB1 have also been described as possible food contaminants, and the very limited data available suggest their significantly higher toxicity compared to FB1. Furthermore, the physicochemical and toxicokinetic properties (e.g., albumin binding) of acyl-FB1 derivatives may show large differences compared to the parent mycotoxin. Therefore, we tested the interactions of FB1, N-palmitoyl-FB1 (N-pal-FB1), 5-O-palmitoyl-FB1 (5-O-pal-FB1), and fumonisin B4 (FB4) with human serum albumin as well as the toxic effects of these mycotoxins on zebrafish embryos were examined. Based on our results, the most important observations and conclusions are the following: (1) FB1 and FB4 bind to albumin with low affinity, while palmitoyl-FB1 derivatives form highly stable complexes with the protein. (2) N-pal-FB1 and 5-O-pal-FB1 likely occupy more high-affinity binding sites on albumin. (3) Among the mycotoxins tested, N-pal-FB1 showed the most toxic effects on zebrafish, followed by 5-O-pal-FB1, FB4, and FB1. (4) Our study provides the first in vivo toxicity data regarding N-pal-FB1, 5-O-pal-FB1, and FB4.


Assuntos
Fumonisinas , Micotoxinas , Animais , Humanos , Fumonisinas/toxicidade , Fumonisinas/metabolismo , Micotoxinas/toxicidade , Peixe-Zebra/metabolismo , Albumina Sérica Humana
19.
Toxins (Basel) ; 15(5)2023 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-37235340

RESUMO

Different mycotoxins in feed lead to combined exposure, increasing adverse effects on animal health. Trichothecene mycotoxins have been associated with inducing oxidative stress, which is neutralized by the glutathione system within the antioxidant defense, depending on the dose and duration of exposure. T-2 toxin, deoxynivalenol (DON), and fumonisin B1 (FB1) are commonly found in feed commodities simultaneously. In the present study, the intracellular biochemical and gene expression changes were investigated in the case of multi-mycotoxin exposure, focusing on certain elements of the glutathione redox system. In a short-term feeding trial, an in vivo study was performed with low (EU-proposed) doses: T-2/HT-2 toxin: 0.25 mg; DON/2-AcDON/15-AcDON.: 5 mg; FB1: 20 mg/kg feed, and high doses (twice the low dose) in laying hens. The multi-mycotoxin exposure affected the glutathione system; GSH concentration and GPx activity was higher in the liver in the low-dose group on day 1 compared to the control. Furthermore, the gene expression of antioxidant enzymes increased significantly on day 1 in both exposure levels compared to the control. The results suggest that when EU-limiting doses are applied, individual mycotoxins may have a synergistic effect in the induction of oxidative stress.


Assuntos
Fumonisinas , Micotoxinas , Toxina T-2 , Animais , Feminino , Toxina T-2/toxicidade , Toxina T-2/metabolismo , Antioxidantes/metabolismo , Galinhas/metabolismo , Fumonisinas/toxicidade , Fumonisinas/metabolismo , Micotoxinas/toxicidade , Micotoxinas/metabolismo , Oxirredução , Glutationa/metabolismo
20.
Int J Mol Sci ; 24(10)2023 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-37240089

RESUMO

The goal of the current study was to examine the effects of prenatal exposure to fumonisins (FBs) on bone properties and metabolism in weaned rat offspring divided into groups intoxicated with FBs at either 0 (the 0 FB group), 60 (the 60 FB group), or 90 mg/kg b.w. 0 (the 90 FB group). Female and male offspring exposed to FBs at a dose of 60 mg/kg b.w. had heavier femora. Mechanical bone parameters changed in a sex and FBs dose-dependent manner. Growth hormone and osteoprotegerin decreased in both sexes, regardless of FBs dose. In males osteocalcin decreased, while receptor activator for nuclear factor kappa-Β ligand increased regardless of FBs dose; while in females changes were dose dependent. Leptin decreased in both male FBs-intoxicated groups, bone alkaline phosphatase decreased only in the 60 FB group. Matrix metalloproteinase-8 protein expression increased in both female FBs-intoxicated groups and decreased in male 90 FB group. Osteoprotegerin and tissue inhibitor of metalloproteinases 2 protein expression decreased in males, regardless of FBs dose, while nuclear factor kappa-Β ligand expression increased only in the 90 FB group. The disturbances in bone metabolic processes seemed to result from imbalances in the RANKL/RANK/OPG and the OC/leptin systems.


Assuntos
Fumonisinas , Osteoprotegerina , Ratos , Masculino , Feminino , Animais , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fumonisinas/toxicidade , Leptina , Ligantes , NF-kappa B/metabolismo , Desenvolvimento Ósseo , Ligante RANK/genética , Ligante RANK/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...