Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Chemosphere ; 303(Pt 1): 134951, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35577126

ABSTRACT

Hexafluoropropylene oxide trimer acid (HFPO-TA) has been used as an alternative of perfluorooctanoic acid (PFOA) in the fluoropolymer industry for several years. HFPO-TA is reported to have high capability of bioaccumulation, widespread environmental distribution, and multiple toxicities. However, its potential toxicity on the intestines and gut microbiota remains unknown. In the present study, male mice were orally exposed to 200 µg/L HFPO-TA for 6 weeks, and after total genomic DNA extraction, 16S rRNA amplicon pyrosequencing was performed. Our results demonstrated that HFPO-TA exposure resulted in the imbalance of cecal microbiota and alterations of cecal microbiota diversity. At the phylum level, the relative abundances of Proteobacteria, Deferribacteres, and Tenericutes increased in mice after exposure to HFPO-TA, while the relative abundances of Verrucomicrobia, Cyanobacteria, and TM7 decreased. At the genus level, the relative abundances of Ver Akkermansia, Pre Prevotella, Lac Coprococcus, Por_Parabacteroides, and Lac Dorea decreased in HFPO-TA exposed mice. Meanwhile, the increased relative abundances of Def_Mucispirillum, Des_Desulfovibrio and Odo Odoribacter were observed in HFPO-TA exposed mice. Additionally, KEGG metabolic pathway analysis revealed that HFPO-TA exposure changed the unsaturated fatty acid synthesis, fatty acid metabolism, glyoxylic acid and dicarboxylic acid metabolism, galactose metabolism pathway and other metabolic pathways. Collectively, all these findings indicate the potential gut toxicity of HFPO-TA and is perceived as a risk of health on gut microbiota. Future investigations should be warranted.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Lipid Metabolism , Male , Mice , Oxides , RNA, Ribosomal, 16S
2.
J Hazard Mater ; 430: 128376, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35158245

ABSTRACT

Hexafluoropropylene oxide trimer acid (HFPO-TA) is reported to have hepatotoxicity, lipotoxicity, and cytotoxicity. In this study, the toxicological effects of HFPO-TA on mitochondrial function and biogenesis were studied. Mice were exposed to drinking water which contained either 2, 20, or 200 µg/L HFPO-TA. Results showed exposure to HFPO-TA induced disadvantageous physiological changes in mice, including increases in liver weight, altered cell morphology, and inflammatory responses. Specifically, exposure to 200 µg/L HFPO-TA increased mitochondria number, relative mitochondrial DNA (mtDNA) content, and mRNA levels of mitochondrial genes encoded by mtDNA. Significant increases in TFAM mRNA and protein levels were also observed. Liver metabolome analysis also showed exposure to 200 µg/L HFPO-TA further enhanced increases in metabolites and altered metabolic pathways that correlated with mitochondrial function, especially the production of ATP. HFPO-TA exposure increased protein expression of mitochondrial complex I-V, and the activities of key enzymes involved in TCA cycle (α-ketoglutarate dehydrogenase, citrate synthase, and succinate dehydrogenase). Furthermore, exposure to 200 µg/L HFPO-TA significantly up-regulating mRNA and protein levels of Opa1, Mfn1, Mfn2, Fis1, and Mff, but did not change Drp1. These findings suggest HFPO-TA could have detrimental effects on health of animals, particularly it was associated with disrupted mitochondrial energy metabolism.


Subject(s)
Fluorocarbons , Animals , Fluorocarbons/toxicity , Liver , Mice , Mitochondria , Oxides
3.
Mol Nutr Food Res ; 66(3): e2100639, 2022 02.
Article in English | MEDLINE | ID: mdl-34847296

ABSTRACT

SCOPE: The impacts of longevity-promoting probiotic Bifidobacterium animalis subsp. lactis LKM512 (LKM512) on metabolic disease remain unclear. Here, the authors aim to explore the potential of LKM512 on the host physiological function and gut microbiota in high-fat diet-induced obese mice. METHODS AND RESULTS: LKM512 are orally administrated for 12 weeks, and the effects of LKM 512 on systemic inflammation and insulin resistance, as well as gut microbiota, are investigated in high-fat (HF) diet-induced obese mice. LKM512 supplementation ameliorates hepatic lipid accumulation, attenuates hepatic and adipose tissue inflammation, and improves intestinal barrier function. These results are associated with improved insulin sensitivity and metabolic endotoxemia. Furthermore, the colonization of LKM512 induces an increase in polyamine metabolism and production, together with significant alternations in the composition and function of gut microbiota in obese mice, which are correlated with these improved metabolic phenotypes in the host. CONCLUSION: The probiotic strain LKM512 may become a promising strategy to improve obesity and related metabolic disorders.


Subject(s)
Bifidobacterium animalis , Gastrointestinal Microbiome , Insulin Resistance , Probiotics , Animals , Bifidobacterium/metabolism , Diet, High-Fat/adverse effects , Feces/microbiology , Inflammation , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Probiotics/pharmacology
4.
Environ Pollut ; 290: 117934, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34416495

ABSTRACT

Hexafluoropropylene oxide dimer acid (HFPO-DA) is the substitute for perfluoro octanoic acid (PFOA), and recently it has been detected in environmental water samples worldwide and has multiple toxicities. However, whether it will affect the intestines and gut microbiota remains unclear. In this study, in order to evaluate the gut toxicity of HFPO-DA in mammals, male mice were orally exposed to 0, 2, 20, 200 µg/L HFPO-DA, respectively, for 6 weeks. Our results showed that HFPO-DA exposure caused colonic inflammation which was coupled with increased TNF-α levels in serum and increased mRNA expression levels of TNF-α, p65, TLR4, MCP-1 of the colon in mice after exposure to 200 µg/L HFPO-DA. We also found that HFPO-DA exposure induced the decreased mRNA expression levels and protein levels of MUC2 and ZO-1, which means the dysfunction of gut barrier in the colon. In the ileum, we found that HFPO-DA exposure induced the increased mRNA expression levels of various inflammatory factors, but no obvious changes was found to barrier function. Additionally, HFPO-DA exposure caused the imbalance of cecal gut microbiota and changes of cecal microbiota diversity. Taken together, all these results indicate the potential gut toxicity of HFPO-DA and is perceived as a major problem of health risk that affects the inflammation, gut barrier dysfunction, and gut microbiota disturbance in mammals.


Subject(s)
Fluorocarbons , Gastrointestinal Microbiome , Microbiota , Animals , Fluorocarbons/toxicity , Male , Mice , Oxides
5.
J Agric Food Chem ; 69(34): 9800-9812, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34404209

ABSTRACT

Aging is the most common cause of several neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease. The pathological hallmarks of age-dependent neuropathology consist of chronic neuroinflammation, oxidative stress, gliosis, learning disability, and cognitive decline. A novel hydrolyzed bioactive peptide mixture extracted from chicken meat, that is, hydrolyzed chicken extract (HCE) has been previously demonstrated to exert neuroprotective effects in rodents and humans. However, the mechanism of HCE on age-related neurological disorders remains unclear. Herein, we aimed to clarify the impact and mechanism of isolated bioactive components (BCs) from HCE on age-dependent neuroinflammation and cognitive impairment in middle-aged mice. We found that both BC and HCE supplementation ameliorated age-induced memory loss, alleviated hippocampal neuroinflammation and oxidative stress, followed by promoting hippocampal neurogenesis in mice. BC and HCE treatment also ameliorated age-dependent morphological anomalies and alleviated microgliosis and astrogliosis. In parallel, BC and HCE treatment showed a significant decrease in the NF-κB p65 and p38 MAPK signaling, which were associated with the enhancement of antioxidative enzymes activities. Furthermore, BC treatment attenuated the neuroinflammatory phenotypes by the decrease in M1-polarized microglia and the increase in M2-polarized microglia in vivo and in vitro. In addition, we found that cyclo(Phe-Phe), one of the cyclopeptides purified from BC, showed notable anti-inflammatory effects in BV2 cells. Taken together, BC might be used as a dietary supplement for alleviating age-dependent neuropathology in middle-aged individuals.


Subject(s)
Cognitive Dysfunction , Microglia , Animals , Chickens , Cognitive Dysfunction/drug therapy , Lipopolysaccharides , Meat , Mice , NF-kappa B , Plant Extracts
6.
Chemosphere ; 282: 130952, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34082316

ABSTRACT

Bisphenol A (BPA) has been found to promote hepatotoxicity, reproductive toxicity, and developmental toxicity. However, the neurotoxicity and mechanism of BPA on cognitive function are still unclear. To that end, eight-week-old adult male and female C57BL/6J mice were exposed to 0.05, 0.5, 5, and 50 mg/kg BPA by dietary supplementation for 22 weeks. BPA exposure impaired learning and memory in male mice, associated with increased neuroinflammation and damaged blood-brain barrier. BPA exposure reduced the tight junctions in the colon, resulting in dysfunction of the gut barrier. The levels of neurotransmitters in the serum, hippocampus, and colon of male mice, including tryptophan, 5-hydroxytryptamine (5-HT) and 5-hydroxyindoleacetic acid, were all decreased by BPA, together with reduced expression of tryptophan and 5-HT metabolism-related genes. Cecal microbiota analysis revealed that the diversity and composition of the microbiota in male mice were markedly altered by BPA, leading to functional profile changes in the microbial community. These results suggest that the neurotoxicity of BPA in male mice may be partly regulated by the interactions of the microbiota-gut-brain axis. However, BPA has little effect on the cognitive function in female mice, which might be caused by the microbial differences and the role of estrogen receptors.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Benzhydryl Compounds , Brain , Cognition , Female , Male , Mice , Mice, Inbred C57BL , Phenols , Serotonin
7.
J Agric Food Chem ; 69(6): 1864-1876, 2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33541082

ABSTRACT

Inflammatory bowel disease (IBD) is associated with acute and chronic inflammation of the gastrointestinal tract and has emerged to be a global disease. Spermidine, a natural polyamine, plays a critical role in maintaining cellular homeostasis. Herein, we investigated the impact and mechanism of spermidine on both dextran sulfate sodium (DSS)- and 2,4,6-trinitrobenzenesulfonic acid solution (TNBS)-induced colitis in mice. We found that spermidine exerted protective effects against acute colitis, evidenced by reduced disease activity index (DAI) and colonic inflammation, increased colonic length, and upregulated tight junction proteins in these two colitis models. Importantly, spermidine exerted significant therapeutic and preventive effects against DSS-induced colitis. Pre- and post-treatment with spermidine reduced the expression of proinflammatory cytokines, phosphorylation of (nuclear factor-κB) NF-κB and (mitogen-activated protein kinase) MAPK, and the activation of F4/80 macrophages and T cells in the colon. Furthermore, spermidine upregulated M2 macrophage markers, whereas it downregulated M1 markers in the inflamed colons. In parallel, spermidine reduced M1 pro-inflammatory markers and enhanced M2 anti-inflammatory genes in RAW264.7 cells. These results revealed that spermidine-ameliorated colitis might be through the regulation of M1/M2 macrophage polarization. In addition, spermidine treatment also alleviated LPS/TNF-α-induced inflammation in Caco-2 cells. Taken together, spermidine prevented and reversed colonic inflammation in colitis mice and might be a promising candidate for IBD intervention.


Subject(s)
Colitis , Spermidine , Animals , Caco-2 Cells , Colitis/chemically induced , Colitis/drug therapy , Colitis/genetics , Colon , Cytokines , Dextran Sulfate/toxicity , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , NF-kappa B
8.
Acta Biochim Biophys Sin (Shanghai) ; 53(4): 419-429, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33637986

ABSTRACT

Neuroinflammation and cognitive decline are the key pathological features in aging that bring detrimental impacts upon quality of life. However, there is no effective anti-aging pharmacological therapy thus far. Dietary supplements in particular essence of chicken (EC) has been found to be an effective remedy for alleviating mental stress and improving memory. In addition, a novel hydrolyzed chicken extract, ProBeptigen/CMI-168 (PB), showed beneficial effects on cognitive ability. However, the antiaging effect and possible mechanism of PB and EC are still unknown. Here, we investigated the antiaging effects of PB and EC on hippocampus-related cognitive decline and neuroinflammation in aged mice. PB and EC were administered for 16 weeks in 10-month-old mice. Both PB and EC treatments ameliorated age-related deterioration of learning and memory, and attenuated oxidative stress and inflammation in the hippocampus. These results were associated with decreased inflammatory cytokine levels and increased neurotransmitter levels in the hippocampus. The overall effects of improving aging-induced cognitive decline were more robust in PB-treated mice, while EC was effective in decreasing oxidative stress and inflammation. Moreover, alterations in the diversity and composition of the gut microbiota in aged mice were also regulated by both PB and EC, which induced distinguished features in the gut microbiota and their related functions. This study showed that PB exerts neuroprotective effects in aged mice, the mechanism of which might be different from that of EC. Therefore, PB has a potential as dietary supplement for ameliorating cognitive dysfunction and neuroinflammation in elderly individuals.


Subject(s)
Aging/metabolism , Cognitive Dysfunction/prevention & control , Dietary Supplements , Hippocampus/metabolism , Neuroprotective Agents/pharmacology , Aging/pathology , Animals , Chickens , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Hippocampus/pathology , Inflammation , Male , Mice , Neuroprotective Agents/chemistry , Species Specificity
9.
Life Sci ; 265: 118739, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33186567

ABSTRACT

AIMS: The therapeutic effects of spermidine on preexisting obese mice have been not fully elucidated. In this study, we assessed the anti-obesity impact of spermidine on high-fat diet (HFD)-induced obese mice. MAIN METHODS: C57BL/6J mice were fed a HFD for 16 weeks to induce obesity, and then treated with or without spermidine via drinking water for additional 8 weeks. The contributions of spermidine in regulating obesity phenotypes and metabolic syndrome were further evaluated. KEY FINDINGS: Spermidine administration lowered fat mass and plasma lipid profile in HFD-induced obese mice without affecting body weight. In addition, spermidine attenuated hepatic steatosis by regulating lipid metabolism and enhancing antioxidant capacity. Moreover, spermidine reduced adipose tissue inflammation by decreasing inflammatory cytokine and chemokines expression, and these results might contributed to the enhanced thermogenic gene expression in brown adipose tissue. Furthermore, spermidine treatment enhanced gut barrier function by up-regulating tight junction- and mucin-related gene expression. SIGNIFICANCE: Spermidine-mediated protective impacts involve the regulation of lipid metabolism, inflammation response, gut barrier function and thermogenesis. These findings demonstrate that spermidine has potentials in treating obesity.


Subject(s)
Fatty Liver/physiopathology , Spermidine/pharmacology , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Animals , Anti-Obesity Agents/pharmacology , Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Fatty Liver/metabolism , Inflammation/metabolism , Lipid Metabolism/drug effects , Lipids , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Spermidine/metabolism , Thermogenesis/drug effects
10.
Metabolism ; 114: 154409, 2021 01.
Article in English | MEDLINE | ID: mdl-33096076

ABSTRACT

BACKGROUND AND OBJECTIVES: The gut-liver axis plays an important role in the pathogenesis of nonalcoholic steatohepatitis (NASH), and increased intestinal permeability causes transfer of endotoxin to the liver, which activates the immune response, ultimately leading to hepatic inflammation. Nuclear receptor Rev-erbα is a critical regulator of circadian rhythm, cellular metabolism, and inflammatory responses. However, the role and mechanism of Rev-erbα in gut barrier function and NASH remain unclear. In the present study, we investigated the involvement of Rev-erbα in the regulation of intestinal permeability and the treatment of NASH. METHODS AND RESULTS: The expression of tight junction-related genes and Rev-erbs decreased in the jejunum, ileum and colon of mice with high cholesterol, high fat diet (CL)-induced NASH. Chromatin immunoprecipitation analysis indicated that REV-ERBα directly bound to the promoters of tight junction genes to regulate intestinal permeability. Pharmacological activation of REV-ERBα by SR9009 protected against lipopolysaccharide-induced increased intestinal permeability both in vitro and in vivo, and these effects were associated with the activation of autophagy and decreased apoptotic signaling of epithelial cells. In addition, the chronopharmacological effects of SR9009 were more potent at Zeitgeber time 0 (ZT0) than at ZT12, which was contrary to the rhythm of Rev-erbs in the gastrointestinal tract. The administration of SR9009 attenuated hepatic lipid accumulation, insulin resistance, inflammation, and fibrosis in mice with CL diet-induced NASH, which might be partly attributed to the enhancement of intestinal barrier function. CONCLUSION: Chronopharmacological activation of REV-ERBα might be a potential strategy to treat intestinal barrier dysfunction-related disorders and NASH.


Subject(s)
Intestinal Mucosa/drug effects , Intestines/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrrolidines/therapeutic use , Receptors, Cytoplasmic and Nuclear/agonists , Repressor Proteins/agonists , Thiophenes/therapeutic use , Tight Junctions/drug effects , Animals , Autophagy/drug effects , Blood Glucose , Caco-2 Cells , Cholesterol/blood , Humans , Insulin/blood , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Permeability/drug effects , Pyrrolidines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Tight Junctions/metabolism , Triglycerides/blood
11.
Gut Microbes ; 12(1): 1-19, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33151120

ABSTRACT

Obesity is associated with impaired intestinal barrier function and dysbiosis of the gut microbiota. Spermidine, a polyamine that acts as an autophagy inducer, has important benefits in patients with aging-associated diseases and metabolic dysfunction. However, the mechanism of spermidine on obesity remains unclear. Here, we show that spermidine intake is negatively correlated with obesity in both humans and mice. Spermidine supplementation causes a significant loss of weight and improves insulin resistance in diet-induced obese (DIO) mice. These effects are associated with the alleviation of metabolic endotoxemia and enhancement of intestinal barrier function, which might be mediated through autophagy pathway and TLR4-mediated microbial signaling transduction. Moreover, spermidine causes the significant alteration of microbiota composition and function. Microbiota depletion compromises function, while transplantation of spermidine-altered microbiota confers protection against obesity. These changes might partly be driven by an SCFA-producing bacterium, Lachnospiraceae NK4A136 group, which was decreased in obese subjects and subsequently increased by spermidine. Notably, the change of Lachnospiraceae NK4A136 group is significantly correlated with enhanced gut barrier function induced by spermidine. Our results indicate that spermidine supplementation may serve as a viable therapy for obesity.


Subject(s)
Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/metabolism , Obesity/drug therapy , Spermidine/pharmacology , Tight Junctions/drug effects , Animals , Autophagy/physiology , Body Weight , Caco-2 Cells , Cell Line, Tumor , Clostridiales/metabolism , Dysbiosis/microbiology , Endotoxemia/drug therapy , Humans , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Obesity/microbiology , Signal Transduction , Tight Junctions/microbiology , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...