Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Biochem Pharmacol ; 214: 115645, 2023 08.
Article in English | MEDLINE | ID: mdl-37321415

ABSTRACT

Cells adapt to stress conditions by increasing glucose uptake as cytoprotective strategy. The efficiency of glucose uptake is determined by the translocation of glucose transporters (GLUTs) from cytosolic vesicles to cellular membranes in many tissues and cells. GLUT translocation is tightly controlled by the activation of Tre-2/BUB2/CDC16 1 domain family 4 (TBC1D4) via its phosphorylation. The mechanisms of glucose uptake under stress conditions remain to be clarified. In this study, we surprisingly found that glucose uptake is apparently increased for the early response to three stress stimuli, glucose starvation and the exposure to lipopolysaccharide (LPS) or deoxynivalenol (DON). The stress-induced glucose uptake was mainly controlled by the increment of ß-catenin level and the activation of RSK1. Mechanistically, ß-catenin directly interacted with RSK1 and TBC1D4, acting as the scaffold protein to recruit activated RSK1 to promote the phosphorylation of TBC1D4. In addition, ß-catenin was further stabilized due to the inhibition of GSK3ß kinase activity which is caused by activated RSK1 phosphorylating GSK3ß at Ser9. In general, this triple protein complex consisting of ß-catenin, phosphorylated RSK1, and TBC1D4 were increased in the early response to these stress signals, and consequently, further promoted the phosphorylation of TBC1D4 to facilitate the translocation of GLUT4 to the cell membrane. Our study revealed that the ß-catenin/RSK1 axis contributed to the increment of glucose uptake for cellular adaption to these stress conditions, shedding new insights into cellular energy utilization under stress.


Subject(s)
GTPase-Activating Proteins , beta Catenin , Animals , Glycogen Synthase Kinase 3 beta/metabolism , GTPase-Activating Proteins/metabolism , beta Catenin/metabolism , Biological Transport , Phosphorylation , Glucose/metabolism , Mammals/metabolism
2.
Food Chem ; 423: 136274, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37159968

ABSTRACT

Deoxynivalenol (DON) is the most frequently contaminated mycotoxin in food and feed worldwide, causing significant economic losses and health risks. Physical and chemical detoxification methods are widely used, but they cannot efficiently and specifically remove DON. In the study, the combination of bioinformatics screening and experimental verification confirmed that sorbose dehydrogenase (SDH) can effectively convert DON to 3-keto-DON and a substance that removes four hydrogen atoms for DON. Through rational design, the Vmax of the mutants F103L and F103A were increased by 5 and 23 times, respectively. Furthermore, we identified catalytic sites W218 and D281. SDH and its mutants have broad application conditions, including temperature ranges of 10-45 °C and pH levels of 4-9. Additionally, the half-lives of F103A at 90 °C (processing temperature) and 30 °C (storage temperature) were 60.1 min and 100.5 d, respectively. These results suggest that F103A has significant potential in the detoxification application of DON.


Subject(s)
Carbohydrate Dehydrogenases , Mycotoxins , Temperature , Food Contamination/analysis
3.
Ecotoxicol Environ Saf ; 259: 115028, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37216862

ABSTRACT

The T-2 toxin and deoxynivalenol (DON), as the most concerned members of trichothecenes, induce cellular stress responses and various toxic effects. Stress granules (SGs) are rapidly formed in response to stress and play an important role in the cellular stress response. However, it is not known whether T-2 toxin and DON induce SG formation. In this study, we found that T-2 toxin induces SG formation, while DON surprisingly suppresses SG formation. Meanwhile, we discovered that SIRT1 co-localized with SGs and regulated SG formation by controlling the acetylation level of the SG nucleator G3BP1. Upon T-2 toxin, the acetylation level of G3BP1 increased, but the opposite change was observed upon DON. Importantly, T-2 toxin and DON affect the activity of SIRT1 via changing NAD+ level in a different manner, though the mechanism remains to be clarified. These findings suggest that the distinct effects of T-2 toxin and DON on SG formation are caused by changes in the activity of SIRT1. Furthermore, we found that SGs increase the cell toxicity of T-2 toxin and DON. In conclusion, our results reveal the molecular regulation mechanism of TRIs on SG formation and provide novel insights into the toxicological mechanisms of TRIs.


Subject(s)
T-2 Toxin , T-2 Toxin/toxicity , DNA Helicases/metabolism , RNA Recognition Motif Proteins , RNA Helicases/metabolism , Sirtuin 1 , Stress Granules , Poly-ADP-Ribose Binding Proteins
4.
Arch Toxicol ; 96(10): 2639-2654, 2022 10.
Article in English | MEDLINE | ID: mdl-35900469

ABSTRACT

Deoxynivalenol (DON) is the most widespread mycotoxin in food and feedstuffs, posing a persistent health threat to humans and farm animals. The susceptibilities of DON vary significantly among animals, following the order of pigs, mice/rats and poultry from the most to least susceptible. However, no study comprehensively disentangles factors shaping species-specific sensitivity. In this review, the toxicokinetics and metabolism of DON are summarized in animals and humans. Generally, DON is fast-absorbed and widely distributed in multiple organs. DON is first enriched in the plasma, liver and kidney and subsequently accumulates in the intestine. There are also key variations among animals. Pigs and humans are highly sensitive to DON, and they have similar absorption rates (1 h < tmax < 4 h), high bioavailability (> 55%) and long clearance time (2 h < t1/2 < 4 h). Also, both species lack detoxification microorganisms and mainly depend on liver glucuronidation and urine excretion. Mice and rats have similar toxicokinetics (tmax < 0.5 h, t1/2 < 1 h). However, a higher proportion of DON is excreted by feces as DOM-1 in rats than in mice, suggesting an important role of gut microbiota in rats. Poultry is least sensitive to DON due to their fast absorption rate (tmax < 1 h), low oral bioavailability (5-30%), broadly available detoxification gut microorganisms and short clearance time (t1/2 < 1 h). Aquatic animals have significantly slower plasma clearance of DON than land animals. Overall, studies on toxicokinetics provide valuable information for risk assessment, prevention and control of DON contamination.


Subject(s)
Mycotoxins , Animals , Biological Availability , Feces , Humans , Mice , Mycotoxins/metabolism , Rats , Swine , Toxicokinetics , Trichothecenes
5.
Eur Respir J ; 60(5)2022 11.
Article in English | MEDLINE | ID: mdl-35618276

ABSTRACT

BACKGROUND: Indoor microbial exposure is associated with asthma, but the health effects of indoor metabolites and chemicals have not been comprehensively assessed. METHODS: We collected classroom dust from 24 junior high schools in three geographically distanced areas in Malaysia (Johor Bahru, Terengganu and Penang), and conducted culture-independent high-throughput microbiome and untargeted metabolomics/chemical profiling. RESULTS: 1290 students were surveyed for asthma symptoms (wheeze). In each centre, we found significant variation in the prevalence of wheeze among schools, which could be explained by personal characteristics and air pollutants. Large-scale microbial variations were observed between the three centres; the potential protective bacteria were mainly from phyla Actinobacteria in Johor Bahru, Cyanobacteria in Terengganu and Proteobacteria in Penang. In total, 2633 metabolites and chemicals were characterised. Many metabolites were enriched in low-wheeze schools, including plant secondary metabolites flavonoids/isoflavonoids (isoliquiritigenin, formononetin, astragalin), indole and derivatives (indole, serotonin, 1H-indole-3-carboxaldehyde), and others (biotin, chavicol). A neural network analysis showed that the indole derivatives were co-occurring with the potential protective microbial taxa, including Actinomycetospora, Fischerella and Truepera, suggesting these microorganisms may pose health effects by releasing indole metabolites. A few synthetic chemicals were enriched in high-wheeze schools, including pesticides (2(3H)-benzothiazolethione), fragrances (2-aminobenzoic acid, isovaleric acid), detergents and plastics (phthalic acid), and industrial materials (4,4-sulfonyldiphenol). CONCLUSIONS: This is the first association study between high-throughput indoor chemical profiling and asthma symptoms. The consistent results from the three centres indicate that indoor metabolites/chemicals could be a better indicator than the indoor microbiome for environmental and health assessments, providing new insights for asthma prediction, prevention and control.


Subject(s)
Air Pollution, Indoor , Asthma , Microbiota , Humans , Air Pollution, Indoor/adverse effects , Air Pollution, Indoor/analysis , Malaysia/epidemiology , Asthma/epidemiology , Students , Dust/analysis , Respiratory Sounds , Bacteria , Indoles
6.
mLife ; 1(2): 183-197, 2022 Jun.
Article in English | MEDLINE | ID: mdl-37731585

ABSTRACT

Gut microbiota composition is suggested to associate with coronavirus disease 2019 (COVID-19) severity, but the impact of gut microbiota on health outcomes is largely unclear. We recruited 81 individuals from Wuhan, China, including 13 asymptomatic infection cases (Group A), 24 COVID-19 convalescents with adverse outcomes (Group C), 31 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) re-positive cases (Group D), and 13 non-COVID-19 healthy controls (Group H). The microbial features of Groups A and D were similar and exhibited higher gut microbial diversity and more abundant short-chain fatty acid (SCFA)-producing species than Group C. Group C was enriched with opportunistic pathogens and virulence factors related to adhesion and toxin production. The abundance of SCFA-producing species was negatively correlated, while Escherichia coli was positively correlated with adverse outcomes. All three groups (A, C, and D) were enriched with the mucus-degrading species Akkermansia muciniphila, but decreased with Bacteroides-encoded carbohydrate-active enzymes. The pathways of vitamin B6 metabolic and folate biosynthesis were decreased, while selenocompound metabolism was increased in the three groups. Specifically, the secondary bile acid (BA) metabolic pathway was enriched in Group A. Antibiotic resistance genes were common among the three groups. Conclusively, the gut microbiota was related to the health outcomes of COVID-19. Dietary supplementations (SCFAs, BA, selenium, folate, vitamin B6) may be beneficial to COVID-19 patients.

7.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502057

ABSTRACT

Cereulide is one of the main food-borne toxins for vomiting synthesized by Bacillus cereus, and it widely contaminates meat, eggs, milk, and starchy foods. However, the toxicological effects and mechanisms of the long-time exposure of cereulide in vivo remain unknown. In this study, oral administration of 50 and 200 µg/kg body weight cereulide in the mice for 28 days caused oxidative stress in liver and kidney tissues and induce abnormal expression of inflammatory factors. In pathogenesis, cereulide exposure activated endoplasmic reticulum stress (ER stress) via the pathways of inositol-requiring enzyme 1α (IRE1α)/Xbox binding protein (XBP1) and PRKR-like ER kinase (PERK)/eukaryotic translation initiation factor 2α (eIF2α), and consequently led to the apoptosis and tissue damages in mouse liver and kidney. In vitro, we confirmed that the accumulation of reactive oxygen species (ROS) caused by cereulide is the main factor leading to ER stress in HepaRG and HEK293T cells. Supplementation of sodium butyrate (NaB) inhibited the activations of IRE1α/XBP1 and PERK/eIF2α pathways caused by cereulide exposure in mice, and reduced the cell apoptosis in liver and kidney. In conclusion, this study provides a new insight in understanding the toxicological mechanism and prevention of cereulide exposure.


Subject(s)
Bacterial Toxins/toxicity , Depsipeptides/toxicity , Kidney/drug effects , Liver/drug effects , Animals , Apoptosis , Cell Line, Tumor , Endoplasmic Reticulum Stress , HEK293 Cells , Humans , Kidney/metabolism , Liver/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , X-Box Binding Protein 1/metabolism , eIF-2 Kinase/metabolism
8.
Environ Pollut ; 288: 117814, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34329069

ABSTRACT

Known as a cause of food poisoning, Bacillus cereus (B. cereus) is widespread in nature. Cereulide, the heat-stable and acid-resistant emetic toxin which is produced by some B. cereus strains, is often associated with foodborne outbreaks, and causes acute emetic toxicity at high dosage exposure. However, the toxicological effect and underlying mechanism caused by chronic low-dose cereulide exposure require to be further addressed. In the study, based on mouse model, cereulide exposure (50 µg/kg body weight) for 28 days induced intestinal inflammation, gut microbiota dysbiosis and food intake reduction. According to the cell models, low dose cereulide exposure disrupted the intestinal barrier function and caused intestinal inflammation, which were resulted from endoplasmic reticulum (ER) stress IRE1/XBP1/CHOP pathway activation to induce cell apoptosis and inflammatory cytokines production. For gut microbiota, cereulide decreased the abundances of Lactobacillus and Oscillospira. Furthermore, cereulide disordered the metabolisms of gut microbiota, which exhibited the inhibitions of butyrate and tryptophan. Interestingly, cereulide exposure also inhibited the tryptophan hydroxylase to produce the serotonin in the gut and brain, which might lead to depression-like food intake reduction. Butyrate supplementation (100 mg/kg body weight) significantly reduced intestinal inflammation and serotonin biosynthesis suppression caused by cereulide in mice. In conclusion, chronic cereulide exposure induced ER stress to cause intestinal inflammation, gut microbiota dysbiosis and serotonin biosynthesis suppression. IRE1 could be the therapeutic target and butyrate supplementation is the potential prevention strategy.


Subject(s)
Gastrointestinal Microbiome , Animals , Bacillus cereus , Depsipeptides , Dysbiosis/chemically induced , Food Contamination/analysis , Inflammation/chemically induced , Mice
9.
Int J Mol Sci ; 21(17)2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32878272

ABSTRACT

Deoxynivalenol (DON)-a type B trichothecene mycotoxin, mainly produced by the secondary metabolism of Fusarium-has toxic effects on animals and humans. Although DON's toxicity in many organs including the adrenal glands, thymus, stomach, spleen, and colon has been addressed, its effects on adipocytes have not been investigated. In this study, 3T3-L1 cells were chosen as the cell model and treated with less toxic doses of DON (100 ng/mL) for 7 days. An inhibition of adipogenesis and decrease in triglycerides (TGs) were observed. DON exposure significantly downregulated the expression of PPARγ2 and C/EBPα, along with that of other adipogenic marker genes in 3T3-L1 cells and BALB/c mice. The anti-adipogenesis effect of DON and the downregulation of the expression of adipogenic marker genes were effectively reversed by PPARγ2 overexpression. The repression of PPARγ2's expression is the pivotal event during DON exposure regarding adipogenesis. DON exposure specifically decreased the di-/trimethylation levels of Histone 3 at lysine 4 in 3T3-L1 cells, therefore weakening the enrichment of H3K4me2 and H3K4me3 at the Pparγ2 promoter and suppressing its expression. Conclusively, DON exposure inhibited PPARγ2 expression via decreasing H3K4 methylation, downregulated the expression of PPARγ2-regulated adipogenic marker genes, and consequently suppressed the intermediate and late stages of adipogenesis. Our results broaden the current understanding of DON's toxic effects and provide a reference for addressing the toxicological mechanism of DON's interference with lipid homeostasis.


Subject(s)
Adipogenesis , Cell Differentiation , Gene Expression Regulation/drug effects , PPAR gamma/antagonists & inhibitors , Trichothecenes/pharmacology , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Protein-alpha/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Male , Mice , Mice, Inbred BALB C , PPAR gamma/genetics , PPAR gamma/metabolism
10.
J Med Microbiol ; 69(5): 748-758, 2020 May.
Article in English | MEDLINE | ID: mdl-32209170

ABSTRACT

Introduction. Neonatal infection with Cronobacter sakazakii can cause severe intestinal damage and necrotizing enterocolitis (NEC). The inflammasome and Toll-like receptors mediate intestinal damage caused by other intestinal pathogens causing NEC, but the exact mechanism is unclear.Aim. We evaluated the molecular mechanisms underlying C. sakazakii-induced NEC.Methodology. The effects of C. sakazakii treatment on two cell lines and a Sprague-Dawley rat model of NEC were evaluated by a cell death assay, western blot and real-time PCR analyses of the NLRP3 inflammasome and downstream factors, and observation of cell and intestinal damage.Results. C. sakazakii caused cellular damage in vitro, as well as intestinal damage in an animal model. NLRP3, caspase-1, TLR4 and MyD88, as well as the downstream factor IL-1ß, were upregulated in C. sakazakii-infected J774A.1 and HT-29 cells. Western blotting showed that C. sakazakii-infected J774A.1 and HT-29 cells and the NEC rat model had higher expression levels of N-terminal gasdermin D (GSDMD) compared with those in the control groups. C. sakazakii and its components promote NF-κB expression via the TLR4/MyD88 signalling pathway, thereby regulating the NLRP3 inflammasome and mediating GSDMD cleavage, resulting in pyroptosis-induced intestinal damage.Conclusion. We found that C. sakazakii upregulates NF-κB via TLR4/MyD88 to promote activation of the NLRP3 inflammasome, leading to the up-regulation of downstream caspase-1, release of IL-1ß, GSDMD-mediated pyroptosis and development of NEC. These findings clarify the mechanisms by which C. sakazakii contributes to NEC.


Subject(s)
Cronobacter sakazakii/physiology , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/microbiology , Gene Expression Regulation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Toll-Like Receptor 4/metabolism , Animals , Animals, Newborn , Cell Cycle/drug effects , Cell Line , Cells, Cultured , Disease Models, Animal , Enterocolitis, Necrotizing/pathology , Host-Pathogen Interactions , Humans , NF-kappa B/metabolism , Pyroptosis , Rats , Signal Transduction , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors
11.
Biochem Pharmacol ; 175: 113868, 2020 05.
Article in English | MEDLINE | ID: mdl-32088259

ABSTRACT

Deoxynivalenol (DON) is the most common mycotoxin in grains, and DON exposure causes gastrointestinal inflammation and systemic immunosuppression. The immunosuppression caused by DON has raised serious concerns about whether it is safe to use probiotics in immunocompromised hosts. Gut microbiota remodeling by Lactobacillus is a potential effective strategy to prevent DON exposure. The athymic nude mice were chose as the model of immunocompromised animals. We tested the effect of the probiotic Lactobacillus rhamnosus GG (LGG) or Lactobacillus acidophilus (LA) supplementation on host protection against DON exposure and the underlying mechanisms in nude mice. DON exposure induced endoplasmic reticulum (ER) stress and impaired intestinal barrier function and microbiota, which were relieved by LGG supplementation but not LA supplementation. LGG supplementation significantly enhanced the intestinal barrier function, increased the body weight and the survival rate in nude mice that exposed to DON for two weeks. Furthermore, LGG supplementation modulated the gut microbiota by increasing the abundance of Bacteroidetes and the levels of the butyrate-producing genes But and Buk to promote butyrate production. Butyrate inhibited the IRE1α/XBP1 signaling pathway to reduce DON-induced intestine injury. In conclusion, LGG supplementation modulated the gut microbiota to promote butyrate production, protecting against DON exposure in nude mice. Both LGG and butyrate show promise for use in protecting against DON exposure.


Subject(s)
Butyrates/metabolism , Gastrointestinal Microbiome/drug effects , Intestinal Diseases/prevention & control , Lacticaseibacillus rhamnosus/growth & development , Probiotics/therapeutic use , Trichothecenes/toxicity , Animals , Food Contamination , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Lacticaseibacillus rhamnosus/enzymology , Male , Mice , Mice, Nude , Permeability , Phosphotransferases (Carboxyl Group Acceptor)/genetics , Phosphotransferases (Carboxyl Group Acceptor)/metabolism , Trichothecenes/metabolism
12.
Toxicol Lett ; 315: 77-86, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31470059

ABSTRACT

T-2 toxin is a major pollutant in crops and feedstuffs. Due to its high toxicity in a variety of organisms, T-2 toxin is of great concern as a threat to humans and to animal breeding. Overexpression of CYP1A1 may contribute to carcinogenesis, and CYP1A1 may be a promising target for the prevention and treatment of human malignancies. Therefore, it is essential to understand the regulatory mechanism by which T-2 toxin induces CYP1A1 expression in human cells. In this study, we confirmed that T-2 toxin (100 ng/mL) induced the expression of CYP1A1 in HepG2 cells through NRF1 and Sp1 bound to the promoter instead of through the well-recognized Aromatic hydrocarbon receptors (AhR). In cells treated with T-2 toxin, Sp1, but not NRF1, was significantly upregulated. However, T-2 toxin apparently promoted the interaction between NRF1 and Sp1 proteins, as revealed by IP analysis. Furthermore, in T-2 toxin-treated HepG2 cells, nuclear translocation of NRF1 was enhanced, while knockdown of Sp1 ablated NRF1 nuclear enrichment. Our results revealed that the upregulation of CYP1A1 by T-2 toxin in HepG2 cells depended on enhanced interaction between Sp1 and NRF1. This finding suggests the tumorigenic features of T-2 toxin might be related to the CYP1A1, which provides new insights to understand the toxicological effect of T-2 toxin.


Subject(s)
Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A1/genetics , Gene Expression Regulation/drug effects , NF-E2-Related Factor 1/genetics , Sp1 Transcription Factor/genetics , T-2 Toxin/toxicity , Up-Regulation/drug effects , Carcinoma/physiopathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Embryo Research , Gene Expression Regulation, Enzymologic , Humans , Kidney , Liver Neoplasms/physiopathology , NF-E2-Related Factor 1/drug effects , NF-E2-Related Factor 1/metabolism , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/metabolism
13.
mSystems ; 4(4)2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31387931

ABSTRACT

Cronobacter sakazakii is an important pathogen associated with the development of necrotizing enterocolitis (NEC), infant sepsis, and meningitis. Several randomized prospective clinical trials demonstrated that oral probiotics could decrease the incidence of NEC. Previously, we isolated and characterized a novel probiotic, Bacteroides fragilis strain ZY-312. However, it remains unclear how ZY-312 protects the host from the effects of C. sakazakii infection. To understand the underlying mechanisms triggering the probiotic effects, we tested the hypothesis that there was cross talk between probiotics/probiotics-modulated microbiota and the local immune system, governed by the permeability of the intestinal mucosa, using in vitro and in vivo models for the intestinal permeability. The probiotic effects of ZY-312 on intestinal epithelial cells were first examined, and the results revealed that ZY-312 inhibited C. sakazakii invasion, C. sakazakii-induced dual cell death (pyroptosis and apoptosis), and epithelial barrier dysfunction in vitro and in vivo The presence of ZY-312 also resulted in decreased expression of an inflammasome (NOD-like receptor family member pyrin domain-containing protein 3 [NLRP3]), caspase-3, and serine protease caspase-1 in a neonatal rat model. Furthermore, ZY-312 significantly modulated the compositions of the intestinal bacterial communities and decreased the relative abundances of Proteobacteria and Gammaproteobacteria but increased the relative abundances of Bacteroides and Bacillus in neonatal rats. In conclusion, our findings have shown for the first time that the probiotic B. fragilis ZY-312 suppresses C. sakazakii-induced NEC by modulating the proinflammatory response and dual cell death (apoptosis and pyroptosis).IMPORTANCE Cronobacter sakazakii is an opportunistic pathogenic bacterium that can cause necrotizing enterocolitis (NEC). However, the mechanism of pathogenicity of C. sakazakii is largely unknown. Here we have now demonstrated that apoptotic and pyroptotic stimuli are effectors of C. sakazakii-induced NEC. Previously, we isolated a novel probiotic strain candidate from fecal samples from healthy infants and characterized it as Bacteroides fragilis strain ZY-312. Functional characterization reveals that ZY-312 inhibited C. sakazakii invasion, restoring epithelial barrier dysfunction, decreasing the expression of inflammatory cytokines, and reducing dual cell death (pyroptosis and apoptosis). Furthermore, the presence of ZY-132 was sufficient to hinder the adverse reaction seen with C. sakazakii in a C. sakazakii-induced NEC model. Taking the results together, our study demonstrated the utility of ZY-312 as a promising probiotic agent for the prevention of NEC.

14.
Toxicology ; 424: 152230, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31170431

ABSTRACT

T-2 toxin is a trichothecene mycotoxin that widely contaminates food and has a variety of toxic effects. However, the underlying mechanism of T-2 toxin on intestinal mucin remains unclear. In present study, human intestinal Caco-2 cells and HT-29 cells were treated with 100 ng/mL T-2 toxin at one-quarter of the IC50 for 24 h, which caused the inhibition of MUC2 and adhesion of E. coli O157:H7. We found T-2 toxin induced endoplasmic reticulum stress and activated the IRE1/XBP1 pathway, which may be related to the inhibition of MUC2. Interestingly, T-2 toxin activated IRE1α to inhibit IRE1ß, which optimized mucin production. Furthermore, overexpression of IRE1ß in the cells apparently alleviated the inhibition of MUC2 caused by T-2 toxin. IRE1α knock-down blocked the down-regulation of IRE1ß and MUC2 induced by T-2 toxin. We revealed the critical role of IRE1α in the inhibition of intestinal mucin. This finding was confirmed in BALB/c mice which were exposed to T-2 toxin (0.5 mg/kg bw) for 4 weeks. T-2 toxin activated the IRE1/XBP1 pathway to disrupt intestinal mucin, which lead to the imbalance of gut microbiota and an increased risk of host infection by E. coli O157:H7. T-2 toxin exposure also increased the expressions of pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α in mice, which might respond to IRE1α activation. Importantly, IRE1α activation was a therapeutic target for intestinal inflammation caused by T-2 toxin. This study provided a new perspective to understand the intestinal toxicity of T-2 toxin.


Subject(s)
Endoribonucleases/metabolism , Mucins/biosynthesis , Protein Serine-Threonine Kinases/metabolism , T-2 Toxin/toxicity , X-Box Binding Protein 1/agonists , Animals , Bacterial Adhesion/drug effects , Caco-2 Cells , Cytokines/biosynthesis , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/drug effects , Enzyme Activation/drug effects , Escherichia coli O157/drug effects , Gastrointestinal Microbiome/drug effects , HT29 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mucin-2/antagonists & inhibitors , Protein Serine-Threonine Kinases/drug effects
15.
FEBS J ; 286(21): 4215-4231, 2019 11.
Article in English | MEDLINE | ID: mdl-31199573

ABSTRACT

Cytochrome P450 1A1 (CYP1A1) is abundant in the kidney, liver, and intestine and is involved in the phase I metabolism of numerous endogenous and exogenous compounds. Therefore, exploring the regulatory mechanism of its basal expression in humans is particularly important to understand the bioactivation of several procarcinogens to their carcinogenic derivatives. Site-specific mutagenesis and deletion of the transcription factor binding site determined the core cis-acting elements in the human CYP1A1 proximal and distal promoter regions. The proximal promoter region [overlapping xenobiotic-responsive element (XRE) and GC box sequences] determined the basal expression of CYP1A1. In human hepatocellular carcinoma cells (HepG2) with aryl hydrocarbon receptor (AhR) or specificity protein 1 (Sp1) knockdown, we confirmed that AhR and Sp1 are involved in basal CYP1A1 expression. In HepG2 cells overexpressing either AhR or Sp1, AhR determined the proximal transactivation of basal CYP1A1 expression. Via DNA affinity precipitation assays and ChIP, we found that AhR bound to the promoter and recruited Sp1 to transactivate CYP1A1 expression. The coordinated interaction between Sp1 and AhR was identified to be DNA mediated. Our work revealed a basal regulatory mechanism of an interesting human gene by which AhR interacts with Sp1 through DNA and recruits Sp1 to regulate basal CYP1A1 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cytochrome P-450 CYP1A1/genetics , Neoplasms/genetics , Receptors, Aryl Hydrocarbon/genetics , Sp1 Transcription Factor/genetics , Carcinogens/toxicity , Gene Expression Regulation/genetics , Hep G2 Cells , Humans , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Metabolic Detoxication, Phase I/genetics , Neoplasms/chemically induced , Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding/genetics , Transcriptional Activation/genetics
16.
Front Microbiol ; 8: 417, 2017.
Article in English | MEDLINE | ID: mdl-28360900

ABSTRACT

Enterohemorrhagic Escherichia coli O157:H7 (EHEC O157:H7) causes hemorrhagic colitis and the formation of characteristic attaching and effacing (A/E) lesions in humans. Given the severe sequelae of EHEC O157:H7 infection, it is critical to develop effective vaccines for human use. However, for achieving this goal many hurdles need to be addressed, such as the type or subset of antigens, adjuvant, and the delivery route. We developed a candidate vaccine by inserting the bivalent antigen espA-Tir-M composed of espA and the Tir central domain into Lactobacillus acidophilus. The recombinant L. acidophilus (LA-ET) was safe in a cell model and excluded EHEC O157:H7 from LoVo cells at rates of nearly 94 and 60% in exclusion and competition assays, respectively. LA-ET inhibited the induction of A/E lesions by EHEC O157:H7 cells in vitro. Oral immunization with LA-ET induced higher levels of specific mucosal and systemic antibody responses in mice. Moreover, LA-ET enhanced interferon-γ and interleukin-4 and -10 production, which was associated with mixed helper T (Th1/Th2) cell responses, and protected against EHEC O157:H7 colonization and infection in mice at a rate of 80%. Histopathological analyses revealed that orally administered LA-ET reduced or inhibited A/E lesions and toxin-induced systemic injury. These findings demonstrate that LA-ET induces both humoral and cellular immune responses in mice and is therefore a promising vaccine against EHEC O157:H7 infection.

17.
Microb Pathog ; 105: 19-24, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28163157

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 causes hemorrhagic colitis and hemolytic uremic syndrome in humans. Due to the risks associated with antibiotic treatment against EHEC O157:H7 infection, vaccines represent a promising method for prevention of EHEC O157:H7 infection. Therefore, we constructed the novel bivalent antigen EspA-Tir-M as a candidate EHEC O157:H7 subunit vaccine. We then evaluated the immunogenicity of this novel EHEC O157:H7 subunit vaccine. Immune responses to the fusion protein administered by intranasal and subcutaneous routes were compared in mice. Results showed higher levels of specific mucosal and systemic antibody responses induced by intranasal as compared to subcutaneous immunization. Intranasal immunization enhanced the concentration of interleukin-4, interleukin-10, and interferon-γ, while subcutaneous immunization enhanced only the latter two. In addition, intranasal immunization protected against EHEC O157:H7 colonization and infection in mice at a rate of 90%.Histopathological analysis revealed that vaccination reduced colon damage, especially when administered intranasally. In contrast, subcutaneous immunization elicited a weak immune response and exhibited a low protection rate. These findings demonstrate that intranasal immunization with the fusion protein induces both humoral and cellular immune (Th1/Th2) responses in mice. The novel EspA-Tir-M novel fusion protein therefore represents a promising subunit vaccine against EHEC O157:H7 infection.


Subject(s)
Escherichia coli Infections/prevention & control , Escherichia coli O157/immunology , Escherichia coli Proteins/immunology , Escherichia coli Vaccines/immunology , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Colitis/microbiology , Colitis/prevention & control , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Vaccines/administration & dosage , Escherichia coli Vaccines/genetics , Female , Immunity, Mucosal , Injections, Subcutaneous , Mice, Inbred BALB C , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
Nan Fang Yi Ke Da Xue Xue Bao ; 35(8): 1079-83, 2015 Aug.
Article in Chinese | MEDLINE | ID: mdl-26277500

ABSTRACT

OBJECTIVE: To investigate the inhibitory effect of Lactobacillus rhamnosus GG ( LGG) against Cronobacter-induced meningitis in neonatal rats. METHODS: The cell adhesion and invasion capacities of Cronobacter were assayed in Caco-2 cells, and the optimal time length and concentration of the bacterium for infection were determined. The suppressive effects of LGG on the adhesion and invasion of Cronobacter in caco-2 cells were tested by competitive and exclusion experiments, and its inhibitory effect against Cronobacter-induced meningitis was evaluated in neonatal rats. RESULTS: Cronobacter showed aggressive adhesion to caco-2 cells with an optimal infection time of 3 h. LGG produced a concentration-dependent inhibition of Cronobacter adhesion and invasion by competing with and excluding the latter for cell adhesion. In neonatal rats, LGG showed an obvious preventive effect and also a moderate therapeutic effect against Cronobacter-induced meningitis. CONCLUSION: LGG can inhibit Cronobacter entry across the intestinal barrier to achieve preventive and therapeutic effects against Cronobacter-induced meningitis.


Subject(s)
Cronobacter/pathogenicity , Enterobacteriaceae Infections/therapy , Lacticaseibacillus rhamnosus , Meningitis, Bacterial/therapy , Probiotics , Animals , Animals, Newborn , Bacterial Adhesion , Caco-2 Cells , Humans , Intestines/microbiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...