RESUMEN
The development of acute respiratory distress syndrome (ARDS) in sepsis is associated with substantial morbidity and mortality. However, the molecular pathogenesis underlying sepsis-induced ARDS remains elusive. Neutrophil heterogeneity and dysfunction contribute to uncontrolled inflammation in patients with ARDS. A specific subset of neutrophils undergoing reverse transendothelial migration (rTEM), which is characterized by an activated phenotype, is implicated in the systemic dissemination of inflammation. Using single-cell RNA sequencing (scRNA-seq), it identified functionally activated neutrophils exhibiting the rTEM phenotype in the lung of a sepsis mouse model using cecal ligation and puncture. The prevalence of neutrophils with the rTEM phenotype is elevated in the blood of patients with sepsis-associated ARDS and is positively correlated with disease severity. Mechanically, scRNA-seq and proteomic analys revealed that inflamed endothelial cell (EC) released extracellular vesicles (EVs) enriched in karyopherin subunit beta-1 (KPNB1), promoting abluminal-to-luminal neutrophil rTEM. Additionally, EC-derived EVs are elevated and positively correlated with the proportion of rTEM neutrophils in clinical sepsis. Collectively, EC-derived EV is identified as a critical regulator of neutrophil rTEM, providing insights into the contribution of rTEM neutrophils to sepsis-associated lung injury.
Asunto(s)
Modelos Animales de Enfermedad , Células Endoteliales , Vesículas Extracelulares , Neutrófilos , Sepsis , Animales , Vesículas Extracelulares/metabolismo , Ratones , Neutrófilos/metabolismo , Sepsis/metabolismo , Sepsis/complicaciones , Sepsis/inmunología , Células Endoteliales/metabolismo , Humanos , Lesión Pulmonar/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Migración Transendotelial y Transepitelial , Ratones Endogámicos C57BL , MasculinoRESUMEN
We develop and validate a method for the rapid determination and identification of 20 ß-lactamase antibiotics traces in goat's milk by combining the solid phase extraction technology with ultra-high performance liquid chromatography-tandem mass spectrometry. Goat milk samples were extracted with acetonitrile twice. The supernatant was then extracted and cleaned by solid-phase extraction using divinylbenzene and N-vinylpyrrolidone copolymer. The method was validated, with limits of quantification (LOQs) of 0.3 µg kg-1, specificities of 1/3 LOQ, linearities (R2) > 0.99, recoveries of 80-110%, repeatabilities <10.0%, and intermediate precisions <10.0%. The developed method was suitable for the routine analysis of ß-lactamase antibiotics residues in goat's milk and was used to test 76 goat milk samples produced in China.
Asunto(s)
Antibacterianos , Cabras , Leche , Extracción en Fase Sólida , Espectrometría de Masas en Tándem , beta-Lactamasas , Animales , Extracción en Fase Sólida/métodos , Leche/química , Espectrometría de Masas en Tándem/métodos , Antibacterianos/análisis , Cromatografía Líquida de Alta Presión/métodos , Límite de Detección , Reproducibilidad de los Resultados , Residuos de Medicamentos/análisis , Contaminación de Alimentos/análisis , Cromatografía Líquida con Espectrometría de MasasRESUMEN
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common life-threatening syndrome with no effective pharmacotherapy. Sepsis-related ARDS is the main type of ARDS and is more fatal than other types. Extracellular vesicles (EVs) are considered novel mediators in the development of inflammatory diseases. Our previous research suggested that endothelial cell-derived EVs (EC-EVs) play a crucial role in ALI/ARDS development, but the mechanism remains largely unknown. Here, we demonstrated that the number of circulating EC-EVs was increased in sepsis, exacerbating lung injury by targeting monocytes and reprogramming them towards proinflammatory macrophages. Bioinformatics analysis and further mechanistic studies revealed that vascular cell adhesion molecule 1 (VCAM1), overexpressed on EC-EVs during sepsis, activated the NF-κB pathway by interacting with integrin subunit alpha 4 (ITGA4) on the monocyte surface, rather than the tissue resident macrophage surface, thereby regulating monocyte differentiation. This effect could be attenuated by decreasing VCAM1 levels in EC-EVs or blocking ITGA4 on monocytes. Furthermore, the number of VCAM1+ EC-EVs was significantly increased in patients with sepsis-related ARDS. These findings not only shed light on a previously unidentified mechanism underling sepsis-related ALI/ARDS, but also provide potential novel targets and strategies for its precise treatment.
Asunto(s)
Lesión Pulmonar Aguda , Vesículas Extracelulares , Monocitos , Sepsis , Molécula 1 de Adhesión Celular Vascular , Humanos , Lesión Pulmonar Aguda/metabolismo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Monocitos/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Sepsis/complicaciones , Sepsis/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismoRESUMEN
BACKGROUND: T helper (Th) cell imbalances have been associated with the pathophysiology of sepsis, including the Th1/Th2 and Th17/T regulatory cells (Treg) paradigms. Cold-inducible RNA-binding protein (CIRP), a novel damage-associated molecular pattern (DAMP) was reported that could induce T cell activation, and skew CD4+ T cells towards a Th1 profile. However, the effect and underlying mechanisms of CIRP on Th17/Treg differentiation in sepsis still remains unknown. METHODS: A prospective exploratory study including patients with sepsis was conducted. Blood samples were collected from patients on days 0, 3 and 7 on admission. The serum CIRP and peripheral blood Treg/Th17 percentage was determined by ELISA and flow cytometry. CD4+ T cells from the spleen and lymph nodes of mice with experimental sepsis were collected after treatment with normal saline (NS), recombinant murine CIRP (rmCIRP) and C23 (an antagonist for CIRP-TLR4) at late stage of sepsis. RNA-seq was conducted to reveal the pivotal molecular mechanism of CIRP on Treg/Th17 differentiation. Naïve CD4+ T cell was isolated from the Tlr4 null and wildtype mice in the presence or absence rmCIRP and C23 to confirmed above findings. RESULTS: A total of 19 patients with sepsis finally completed the study. Serum CIRP levels remained high in the majority of patients up to 1 week after admittance was closely associated with high Treg/Th17 ratio of peripheral blood and poor outcome. A univariate logistic analysis demonstrated that higher CIRP concentration at Day 7 is an independent risk factor for Treg/Th17 ratio increasing. CIRP promotes Treg development and suppresses Th17 differentiation was found both in vivo and in vitro. Pretreated with C23 not only alleviated the majority of negative effect of CIRP on Th17 differentiation, but also inhibited Treg differentiation, to some extent. Tlr4 deficiency could abolish almost all downstream effects of rmCIRP. Furthermore, IL-2 is proved a key downstream molecules of the effect CIRP, which also could amplify the activated CD4+ T lymphocytes. CONCLUSIONS: Persistent high circulating CIRP level may lead to Treg/Th17 ratio elevated through TLR4 and subsequent active IL-2 signaling which contribute to immunosuppression during late phases of sepsis.
Asunto(s)
Factores de Transcripción Forkhead , Interleucina-2 , Ratones Endogámicos C57BL , Proteínas de Unión al ARN , Sepsis , Transducción de Señal , Linfocitos T Reguladores , Células Th17 , Receptor Toll-Like 4 , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Diferenciación Celular , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Interleucina-2/metabolismo , Ratones Noqueados , Estudios Prospectivos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Sepsis/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genéticaRESUMEN
Porcine epidemic diarrhea virus (PEDV), a member of the family Coronaviridae, causes acute diarrhea, vomiting, dehydration, and high mortality in newborn piglets, and has caused significant economic losses in the pig industry. There are currently no specific drugs available to treat PEDV. Viruses depend exclusively on the cellular machinery to ensure an efficient replication cycle. In the present study, we found that small-molecule RAF265, an anticancer drug that has been shown to be a potent inhibitor of RAF, reduced viral loads of PEDV by 4 orders of magnitude in Vero cells, and protected piglets from virus challenge. RAF265 reduced PEDV production by mediating cytoskeleton arrangement and targeting the host cell's translation machinery. Treatment with RAF265 inhibited viral entry of PEDV S-glycoprotein pseudotyped viral vector particle (PEDV-pp), at half maximal effective concentrations (EC50) of 79.1 nM. RAF265 also presented potent inhibitory activity against viral infection by SARS-CoV-2-pp and SARS-CoV-pp. The present work may provide a starting point for further progress toward the development of antiviral strategies effective against coronavirus PEDV.
Asunto(s)
COVID-19 , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Chlorocebus aethiops , Animales , Porcinos , Células Vero , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéuticoRESUMEN
Aflatoxin M1 (AFM1), the only toxin with maximum residue levels in milk, has adverse effects on the intestinal barrier, resulting in intestinal inflammatory disease. Lactoferrin (LF), one of the important bioactive proteins in milk, performs multiple biological functions, but knowledge of the protective effects of LF on the compromised intestinal barrier induced by AFM1 has not been investigated. In the present study, results using Balb/C mice and differentiated Caco-2 cells showed that LF intervention decreased AFM1-induced increased intestinal permeability, improved the protein expression of claudin-3, occludin and ZO-1, and repaired the injured intestinal barrier. The transcriptome and proteome were used to clarify the underlying mechanisms. It was found that LF reduced the intestinal barrier dysfunction caused by AFM1 and was associated with intestinal cell survival related pathways, such as cell cycle, apoptosis and MAPK signaling pathway and intestinal integrity related pathways including endocytosis, tight junction, adherens junction and gap junction. The cross-omics analysis suggested that insulin receptor (INSR), cytoplasmic FMR1 interacting protein 2 (CYFIP2), dedicator of cytokinesis 1 (DOCK1) and ribonucleotide reductase regulatory subunit M2 (RRM2) were the potential key regulators as LF repaired the compromised intestinal barrier. These findings indicated that LF may be an alternative treatment for the compromised intestinal barrier induced by AFM1.
Asunto(s)
Aflatoxina M1/toxicidad , Intestinos/patología , Lactoferrina/farmacología , Animales , Peso Corporal/efectos de los fármacos , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Claudina-3/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Intestinos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ocludina/metabolismo , Proteoma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcriptoma/genética , Proteína de la Zonula Occludens-1/metabolismoRESUMEN
Aflatoxin M1 (AFM1) is the only mycotoxin with maximum residue limit in milk, which may result in serious human diseases. On the contrary, lactoferrin (Lf) is an active protein with multiple functions. Studies have confirmed that Lf has a powerful potential to protect the intestines, but the influence of Lf on mycotoxins is not clear. This study aims to explore whether Lf can protect the cytotoxicity induced by AFM1, and determine the underlying mechanisms in human normal colonic epithelial NCM460 cells. The results indicated that AFM1 decreased the cell viability, and increased the levels of apoptosis and autophagy of NCM460 cells. Lf can alleviate the cytotoxicity induced by AFM1 through enhancing cell viability, significantly down-regulated the expression of apoptotic genes and proteins (BAX, caspase3, caspase9, caspase3, and caspase9), and regulated the gene and protein expression of autophagy factors (Atg5, Atg7, Atg12, Beclin1, ULK1, ULK2, LC3, and p62). Furthermore, interference of the key gene Atg5 of autophagy can reduce AFM1-induced apoptosis, which is consistent with the role of Lf, implying that Lf may protect AFM1-induced intestinal injury by inhibiting excessive autophagy-mediated apoptosis. Taken together, our data indicated that Lf has a mitigating effect on apoptosis induced by AFM1 through the autophagy pathway.
RESUMEN
BACKGROUND: Previous studies on the effects of mycotoxins have solely focused on their biochemical profiles or products in dairy ruminants. Changes in metabolism that occur after exposure to mycotoxins, as well as biochemical changes, have not been explored. METHODS: We measured the biochemical and metabolic changes in dairy cows after exposure to mycotoxins using biochemical analyses and nuclear magnetic resonance. Twenty-four dairy cows were randomly assigned to three different treatment groups. Control cows received diets with 2 kg uncontaminated cottonseed. Cows in the 50% replacement group received the same diet as the control group, but with 1 kg of uncontaminated cottonseed and 1 kg of cottonseed contaminated with mycotoxins. Cows in the 100% replacement group received the same diet as the control, but with 2 kg contaminated cottonseed. RESULTS: The results showed that serum γ-glutamyl transpeptidase and total antioxidant capacities were significantly affected by cottonseed contaminated with mycotoxins. There were also significant differences in isovalerate and NH3-N levels, and significant differences in the eight plasma metabolites among the three groups. These metabolites are mainly involved in amino acid metabolism pathways. Therefore, the results suggest that amino acid metabolism pathways may be affected by mycotoxins exposure.
RESUMEN
Milk yield and several components of milk that are affected by physiological factors have been widely investigated. However, the effects of lactation stage and parity on bovine milk whey proteins have not been well elucidated. To aid in unraveling the proteome profile and exploring the protein biosynthesis of mammary glands, a label-free proteomic approach was used to characterize whey proteomes depending on the lactation stage and parity of dairy cows. The results of this study show that the abundances of several proteins, such as early lactation protein, syntenin, and heparanase, were associated with specific stages of the lactation cycle; this was evidenced by a principal component analysis. In addition, several proteins, such as hemoglobin subunits beta and alpha, ß-lactoglobulin, CD320, and apolipoprotein E, corresponded to the parity of the dairy cows and were herein considered as useful biomarkers to distinguish different parities. Most of the differentially expressed proteins from specific lactation stages and parity milk groups were annotated in the response to stimulus and protein metabolic processes. The findings reveal that developmental changes in whey proteomes correspond to lactation stages and parities, which in turn provides new insight into the underlying implications of the production of specific proteins to meet the health benefits of offspring and host, and allow us to explore the mechanisms of protein biosynthesis in mammary glands associated with physiological changes in dairy cows.
Asunto(s)
Lactancia/metabolismo , Paridad , Proteoma/metabolismo , Proteómica/métodos , Proteína de Suero de Leche/metabolismo , Animales , Bovinos , FemeninoRESUMEN
The toxicity and related mechanisms of aflatoxin B1 (AFB1) and aflatoxin M1 (AFM1) in the mouse kidney were studied, and the role of l-proline in alleviating kidney damage was investigated. In a 28-day toxicity mouse model, thirty mice were divided into six groups: control (without treatment), l-proline group (10 g/kg body weight (b.w.)), AFB1 group (0.5 mg/kg b.w.), AFM1 (3.5 mg/kg b.w.), AFB1 + l-proline group and AFM1 + l-proline group. Kidney index and biochemical indicators were detected, and pathological staining was observed. Using a human embryonic kidney 293 (HEK 293) cell model, cell apoptosis rate and apoptotic proteins expressions were detected. The results showed that AFB1 and AFM1 activated pathways related with oxidative stress and caused kidney injury; l-proline significantly alleviated abnormal expressions of biochemical parameters and pathological kidney damage, as well as excessive cell apoptosis in the AF-treated models. Moreover, proline dehydrogenase (PRODH) was verified to regulate the levels of l-proline and downstream apoptotic factors (Bax, Bcl-2, and cleaved Caspase-3) compared with the control (p < 0.05). In conclusion, l-proline could protect mouse kidneys from AFB1 and AFM1 through alleviating oxidative damage and decreasing downstream apoptosis, which deserves further research and development.
Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Aflatoxina B1/toxicidad , Aflatoxina M1/toxicidad , Estrés Oxidativo/efectos de los fármacos , Prolina/uso terapéutico , Sustancias Protectoras/uso terapéutico , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Prolina/farmacología , Prolina Oxidasa/metabolismo , Sustancias Protectoras/farmacología , Pruebas de Toxicidad SubagudaRESUMEN
Aflatoxin M1 (AFM1) and ochratoxin A (OTA), which widely coexist in milk, may pose a serious threat to human health. Mucin is a major component of the intestinal mucus layer, which plays an important role in maintaining intestinal mucosal homeostasis. However, the effect of mycotoxins AFM1 and OTA on intestinal mucin production is still not clear. This study aimed to investigate individual and interactive effects of mycotoxins AFM1 and OTA on the intestinal barrier and the mRNA expression of intestinal mucin (MUC2, MUC5AC and MUC5B) and on protein production in Caco-2/HT29-MTX cultures after 48 h of exposure. Our results show that individual mycotoxins and their mixtures significantly reduced intestinal cell viability and transepithelial electrical resistance (TEER) values, as well as significantly altered intestinal mucin mRNA expression and protein abundance. Moreover, OTA showed toxicity similar to AFM1 in cell viability and TEER value at the same concentration. When the two mycotoxins acted in combination, the synergistic effects observed in the assessment of cell viability and protein abundance in all mono- and co-cultures. In general, this study provides evidence that AFM1 and OTA can damage the intestine, and it contributes to optimized maximum permissible limits of mycotoxins in milk.
Asunto(s)
Aflatoxina M1/toxicidad , Mucina 5AC/genética , Mucina 2/genética , Mucina 5B/genética , Ocratoxinas/toxicidad , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Células HT29 , Humanos , Mucina 5AC/metabolismo , Mucina 2/metabolismo , Mucina 5B/metabolismo , ARN Mensajero/metabolismoRESUMEN
Aflatoxin M1 (AFM1), ochratoxin A (OTA), and zearalenone (ZEA) are mycotoxins commonly found in milk. Mycotoxin contamination has caused food safety concerns worldwide since most of the toxic effects in humans are serious. The combined toxic effects of these mycotoxins on intestinal epithelial cells have not been reported. Herein, we investigated the combined effects of AFM1, OTA, and ZEA on intestinal integrity and define the underlying mechanisms(s) of their effects in Caco-2/HT29-MTX co-cultures. Our results showed that the mixtures of AFM1 + OTA, AFM1 + ZEA, and AFM1 + ZEA + OTA significantly increased epithelial permeability. Immunofluorescence analysis and transmission electron microscopy revealed that mycotoxins altered TJ proteins morphology and disrupted their structures. Also, the present study showed that mixtures of mycotoxins significantly modulated MUC5AC and MUC5B mRNA levels and protein secretion. This study demonstrated that the effects of mixtures of mycotoxins on intestinal barrier function were more significant than AFM1 alone. More importantly, the damage of intestinal integrity caused by mycotoxins was correlated to the change of the TJ proteins location and the decrease of mucin secretion. Mixtures of AFM1, OTA, and ZEA in food might pose a health risk to consumers, particularly in children, and toxin risks should be considered.
Asunto(s)
Aflatoxina M1/toxicidad , Mucosa Intestinal/efectos de los fármacos , Mucinas/metabolismo , Ocratoxinas/toxicidad , Zearalenona/toxicidad , Células CACO-2 , Técnicas de Cocultivo , Contaminación de Alimentos , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Mucina 5AC/análisis , Mucina 5AC/genética , Mucina 2/análisis , Mucina 2/genética , Mucina 5B/análisis , Mucina 5B/genética , Mucinas/genética , Permeabilidad , ARN Mensajero/análisis , Proteínas de Uniones Estrechas/análisisRESUMEN
The milk fat globule membrane (MFGM) plays an important role in stabilizing fat in the aqueous phase, and the components of this membrane are involved in multiple biological functions. Here, we investigated changes in the protein composition of the MFGM fraction between raw and heated whole milk using a label free proteomic approach. In total, 612 MFGM-related proteins were identified in all groups. Compared with raw milk, the number of proteins that were not identified in the MFGM fraction was increased from pasteurized milk to ultra-high-temperature milk, whereas the number of milk proteins incorporated into the MFGM fraction was similar among heated groups. The abundances of several milk proteins (ß-lactoglobulin and ß-casein) were increased in the heated milk groups in a temperature-dependent manner. From our functional analysis, proteins that were not identified in the MFGM fraction of heated milk were mainly associated with multiple biological functions. These findings provided novel insights into the effects of heat procedures on MFGM protein components and their potential physiological functions, thereby yielding data on the appropriate heating procedures to use for raw milk.
Asunto(s)
Glucolípidos/química , Glicoproteínas/química , Calor , Proteínas de la Leche/química , Leche/química , Proteómica/métodos , Animales , Caseínas/análisis , Bovinos , Manipulación de Alimentos/métodos , Glucolípidos/fisiología , Glicoproteínas/fisiología , Lactoglobulinas/análisis , Gotas Lipídicas , Proteínas de la Leche/análisis , PasteurizaciónRESUMEN
Aflatoxin M1 (AFM1) is a common mycotoxin in dairy milk, and it is typically concurrently present with other mycotoxins that may represent a threat to food safety. However, knowledge of how AFM1, alone or in combination with other mycotoxins, may affect human intestinal epithelial integrity remain to be established. We employed transcriptome and proteome analysis integrated with biological validation to reveal the molecular basis underlining the effect of exposure to AFM1, ochratoxin A (OTA), or both on the intestinal epithelial integrity of differentiated Caco-2 cells. Exposure to 4 µg/mL of OTA was found to disrupt human gut epithelial integrity, whereas 4 µg/mL of AFM1 did not. The integrated transcriptome and proteome analysis of AFM1 and OTA, alone or in combination, indicate the synergistic effect of the two mycotoxins in disrupting intestinal integrity. This effect was mechanistically linked to a broad range of pathways related to intestinal integrity enriched by down-regulated genes and proteins, associated with focal adhesion, adheren junction, and gap junction pathways. Furthermore, the cross-omics analysis of mixed AFM1 and OTA compared to OTA alone suggest that kinase family members, including myosin light-chain kinase, mitogen-activated protein kinases, and protein kinase C, are the potential key regulators in modulating intestinal epithelial integrity. These findings provide novel insight into the synergistic detrimental role of multiple mycotoxins in disrupting intestinal integrity and, therefore, identify potential targets to improve milk safety related to human health.
Asunto(s)
Aflatoxina M1/toxicidad , Adhesiones Focales/efectos de los fármacos , Ocratoxinas/toxicidad , Proteoma/genética , Transcriptoma , Uniones Adherentes/efectos de los fármacos , Células CACO-2 , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Uniones Comunicantes/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Mapas de Interacción de Proteínas , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteoma/clasificación , Proteoma/metabolismoRESUMEN
This survey was performed to investigate the occurrence of aflatoxin M1 (AFM1) contamination of raw milk from manufacturers of infant milk powder in China. A total of 1207 raw milk samples were collected overall from four seasons of 2016 in Northeast China, Northwest China, Northern China, and Central China (11 provinces and one municipality). Results showed that 56 of the 1207 raw milk samples (4.64%) were positive for AFM1, which were obtained from Heilongjiang (two samples), Gansu (one sample), Shaanxi (46 samples), Beijing (one sample), and Hunan (six samples) provinces. None of the raw milk samples from manufacturers of infant milk powder exceeded the Chinese limit (62.5 ng/L) in 2016. Only a few raw milk samples were not suitable for use in infant milk according to EU (European Union) or U.S. infant milk limits. Furthermore, based on this survey and previous studies, it is particularly important to avoid AFM1 contamination in raw milk during the winter.
Asunto(s)
Aflatoxina M1/análisis , Contaminación de Alimentos/estadística & datos numéricos , Fórmulas Infantiles/química , Leche/química , Venenos/análisis , Animales , China , Contaminación de Alimentos/análisis , Humanos , Lactante , Fórmulas Infantiles/análisis , PolvosRESUMEN
Aflatoxins, including aflatoxin B1 (AFB1) and M1 (AFM1), are natural potent carcinogens produced by Aspergillus spp. These compounds, which can often be detected in dairy foods, can cause diseases in human beings. However, the molecular mechanisms involved in cytotoxicity, as well as methods for intervention, remain largely unexplored. For example, it is unclear whether lactoferrin (LF), a major antioxidant in milk, can inhibit the cytotoxicity of AFB1 and AFM1. In this study, we assessed AFB1- and AFM1-induced cell toxicity by measuring cell viability, membrane permeability, and genotoxicity, and then investigated the ability of LF to protect cells against AFB1 and AFM1. In Caco-2, HEK, Hep-G2, and SK-N-SH cells, 4⯵g/mL AFB1 or AFM1 significantly inhibited cell growth, increased the level of lactate dehydrogenase, induced genetic damage, and increased the levels of signal-regulated kinase (ERK1/2) and c-Jun N-terminal kinase (JNK) (pâ¯<â¯0.05). AFB1 was more genotoxic than AFM1 in all four cell lines, especially in Hep-G2. In Caco-2, Hep-G2, and SK-N-SH, incubation of AF-treated cells with 1000⯵g/mL LF significantly decreased cytotoxicity, oxidation level, DNA damage, and levels of ERK1/2 and JNK (pâ¯<â¯0.05). Our data demonstrate that AFB1 or AFM1 induced cytotoxicity and DNA damage in these four cell lines, and that LF alleviated toxicity by decreasing oxidative stress mediated by mitogen-activated protein kinase pathways.
Asunto(s)
Aflatoxina B1/toxicidad , Aflatoxina M1/toxicidad , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Lactoferrina/farmacología , Línea Celular Tumoral , HumanosRESUMEN
The object of this study is to analyze the levels of seven toxic elements residues in raw bovine milk in China and assess the potential health risk of those residues. The 178 raw bovine milk samples were collected from eight main milk-producing provinces and from three types of milk stations in China, and were analyzed for arsenic (As), lead (Pb), cadmium (Cd), chromium (Cr), mercury (Hg), aluminum (Al), and nickel (Ni) using inductively coupled plasma-mass spectrometry (ICP-MS). Al, Pb, Hg, Ni, Cr, and As were detected in 47.8, 29.2, 28.1, 23.6, 12.4, and 9.0% of total milk samples, respectively, and Cd were not detected in all samples. The raw bovine milk samples with high levels of toxic elements were found in industrial areas, such as Heilongjiang and Shanxi. Nemerow pollution index analysis showed that the levels were lower in the samples from the processing plants than that from the large-scale farms and small farm cooperatives. The margin of exposure (MOE) values suggest that the levels of As, Pb, Hg, Cr, Al, and Ni in the raw milk samples are not causing a health risk for Chinese consumers, including adults and children. Nevertheless, the risk of Pb for infant and young children was more serious than adult.
Asunto(s)
Análisis de los Alimentos , Metales Pesados/análisis , Leche/química , Adulto , Animales , Bovinos , Niño , Preescolar , Humanos , Espectrometría de Masas , Medición de RiesgoRESUMEN
Aflatoxin M1 (AFM1) and ochratoxin A (OTA) are mycotoxins commonly found in milk; however, their effects on intestinal epithelial cells have not been reported. In the present study, we show that AFM1 (0.12 and 12 µM) and OTA (0.2 and 20 µM) individually or collectively increased the paracellular flux of lucifer yellow and fluorescein isothiocyanate (FITC)-dextrans (4 and 40 kDa) and decreased transepithelial electrical resistance values in differentiated Caco-2 cells after 48 h of exposure, indicating increased epithelial permeability. Immunoblotting and immunofluorescent analysis revealed that AFM1, OTA, and their combination decreased the expression levels of tight junction (TJ) proteins and disrupted their structures, namely, claudin-3, claudin-4, occludin, and zonula occludens-1 (ZO-1), and p44/42 mitogen-activated protein kinase (MAPK) partially involved in the mycotoxins-induced disruption of intestinal barrier. The effects of a combination of AFM1 and OTA on intestinal barrier function were more significant (p < 0.05) than those of AFM1 and OTA alone, yielding additive or synergistic effects. The additive or synergistic effects of AFM1 and OTA on intestinal barrier function might affect human health, especially in children, and toxin risks should be considered.
Asunto(s)
Aflatoxina M1/farmacología , Mucosa Intestinal/efectos de los fármacos , Ocratoxinas/farmacología , Células CACO-2 , Diferenciación Celular , Dextranos/farmacología , Sinergismo Farmacológico , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacología , Colorantes Fluorescentes/farmacología , Humanos , Mucosa Intestinal/metabolismo , Isoquinolinas/farmacología , Permeabilidad/efectos de los fármacos , Proteínas de Uniones Estrechas/metabolismoRESUMEN
In recent years, high levels of hormone residue in food, capable of damaging the health of consumers, have been recorded frequently. In this study, 195 raw milk samples were obtained from Tangshan City, China, and the concentrations of 22 steroid hormones were measured by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Cortisol was detected in 12.5% of raw milk samples (mean 0.61 µg/kg; range: Asunto(s)
Contaminación de Alimentos/análisis
, Leche/química
, Esteroides/aislamiento & purificación
, Animales
, Niño
, Preescolar
, China
, Cromatografía Líquida de Alta Presión
, Humanos
, Lactante
, Medición de Riesgo
, Encuestas y Cuestionarios
, Espectrometría de Masas en Tándem/métodos
RESUMEN
The purposes of this study were to investigate the systemic and characteristic metabolites in the serum of dairy goats induced by aflatoxin B1 (AFB1) exposure and to further understand the endogenous metabolic alterations induced by it. A nuclear magnetic resonance (NMR)-based metabonomic approach was used to analyse the metabolic alterations in dairy goats that were induced by low doses of AFB1 (50 µg/kg DM). We found that AFB1 exposure caused significant elevations of glucose, citrate, acetate, acetoacetate, betaine, and glycine yet caused reductions of lactate, ketone bodies (acetate, ß-hydroxybutyrate), amino acids (citrulline, leucine/isoleucine, valine, creatine) and cell membrane structures (choline, lipoprotein, N-acetyl glycoproteins) in the serum. These data indicated that AFB1 caused endogenous metabolic changes in various metabolic pathways, including cell membrane-associated metabolism, the tricarboxylic acid cycle, glycolysis, lipids, and amino acid metabolism. These findings provide both a comprehensive insight into the metabolic aspects of AFB1-induced adverse effects on dairy goats and a method for monitoring dairy animals exposed to low doses of AFB1.