Metabolomic, Lipidomic, Transcriptomic, and Metagenomic Analyses in Mice Exposed to PFOS and Fed Soluble and Insoluble Dietary Fibers

Background: Perfluorooctane sulfonate (PFOS) is a persistent environmental pollutant that has become a significant concern around the world. Exposure to PFOS may alter gut microbiota and liver metabolic homeostasis in mammals, thereby increasing the risk of cardiometabolic diseases. Diets high in soluble fibers can ameliorate metabolic disease risks. Objectives: We aimed to test the hypothesis that soluble fibers (inulin or pectin) could modulate the adverse metabolic effects of PFOS by affecting microbe-liver metabolism and interactions. Methods: Male C57BL/6J mice were fed an isocaloric diet containing different fibers: a) inulin (soluble), b) pectin (soluble), or c) cellulose (control, insoluble). The mice were exposed to PFOS in drinking water (3μg/g per day) for 7 wk. Multi-omics was used to analyze mouse liver and cecum contents. Results: In PFOS-exposed mice, the number of differentially expressed genes associated with atherogenesis and hepatic hyperlipidemia were lower in those that were fed soluble fiber than those fed insoluble fiber. Shotgun metagenomics showed that inulin and pectin protected against differences in microbiome community in PFOS-exposed vs. control mice. It was found that the plasma PFOS levels were lower in inulin-fed mice, and there was a trend of lower liver accumulation of PFOS in soluble fiber-fed mice compared with the control group. Soluble fiber intake ameliorated the effects of PFOS on host hepatic metabolism gene expression and cecal content microbiome structure. Discussions: Results from metabolomic, lipidomic, and transcriptomic studies suggest that inulin- and pectin-fed mice were less susceptible to PFOS-induced liver metabolic disturbance, hepatic lipid accumulation, and transcriptional changes compared with control diet-fed mice. Our study advances the understanding of interaction between microbes and host under the influences of environmental pollutants and nutrients. The results provide new insights into the microbe-liver metabolic network and the protection against environmental pollutant-induced metabolic diseases by high-fiber diets. https://doi.org/10.1289/EHP11360

[1]  Yuanxiang Jin,et al.  Aberrant hepatic lipid metabolism associated with gut microbiota dysbiosis triggers hepatotoxicity of novel PFOS alternatives in adult zebrafish. , 2022, Environment international.

[2]  E. Hatano,et al.  Propionic Acid, Induced in Gut by an Inulin Diet, Suppresses Inflammation and Ameliorates Liver Ischemia and Reperfusion Injury in Mice , 2022, Frontiers in Immunology.

[3]  G. Bothun,et al.  Replacement per- and polyfluoroalkyl substances (PFAS) are potent modulators of lipogenic and drug metabolizing gene expression signatures in primary human hepatocytes. , 2022, Toxicology and applied pharmacology.

[4]  Yawei Wang,et al.  Effect of Enterohepatic Circulation on the Accumulation of Per- and Polyfluoroalkyl Substances: Evidence from Experimental and Computational Studies. , 2022, Environmental science & technology.

[5]  Shaoqi Yang,et al.  Inulin activates FXR-FGF15 signaling and further increases bile acids excretion in non-alcoholic fatty liver disease mice. , 2022, Biochemical and biophysical research communications.

[6]  Yung-Fu Chang,et al.  Gut microbiota disturbance exaggerates battery wastewater-induced hepatotoxicity through a gut-liver axis , 2021, Science of The Total Environment.

[7]  J. Peters,et al.  The role of mouse and human peroxisome proliferator-activated receptor-α in modulating the hepatic effects of perfluorooctane sulfonate in mice. , 2021, Toxicology.

[8]  M. Orešič,et al.  Exposure to environmental contaminants is associated with altered hepatic lipid metabolism in non-alcoholic fatty liver disease. , 2021, Journal of hepatology.

[9]  C. Fava,et al.  Exposure to Perfluoroalkyl Chemicals and Cardiovascular Disease: Experimental and Epidemiological Evidence , 2021, Frontiers in Endocrinology.

[10]  J. Xia,et al.  MetaboAnalyst 5.0: narrowing the gap between raw spectra and functional insights , 2021, Nucleic Acids Res..

[11]  Yangyang Zhang,et al.  Distribution of perfluorooctane sulfonate in mice and its effect on liver lipidomic. , 2021, Talanta.

[12]  P. O’Toole,et al.  Dietary Fibre Modulates the Gut Microbiota , 2021, Nutrients.

[13]  Wei Wei,et al.  Perfluorooctanesulfonic acid (PFOS) thwarts the beneficial effects of calorie restriction and Metformin. , 2021, Toxicological sciences : an official journal of the Society of Toxicology.

[14]  H. Moseley,et al.  Untargeted Stable Isotope Probing of the Gut Microbiota Metabolome Using 13C-Labeled Dietary Fibers. , 2021, Journal of proteome research.

[15]  R. Choi,et al.  Ceramides and other sphingolipids as drivers of cardiovascular disease , 2021, Nature Reviews Cardiology.

[16]  A. Baccarelli,et al.  Hallmarks of environmental insults , 2021, Cell.

[17]  F. Foufelle,et al.  Roles of Ceramides in Non-Alcoholic Fatty Liver Disease , 2021, Journal of clinical medicine.

[18]  B. Hennig,et al.  Co-exposure to PCB126 and PFOS increases biomarkers associated with cardiovascular disease risk and liver injury in mice. , 2020, Toxicology and applied pharmacology.

[19]  M. Weickert,et al.  The Health Benefits of Dietary Fibre , 2020, Nutrients.

[20]  Marisa Pfohl,et al.  Perfluorooctanesulfonic acid (PFOS) and perfluorohexanesulfonic acid (PFHxS) alter the blood lipidome and the hepatic proteome in a murine model of diet-induced obesity. , 2020, Toxicological sciences : an official journal of the Society of Toxicology.

[21]  F. Akhlaghi,et al.  Perfluorooctanesulfonic acid (PFOS) administration shifts the hepatic proteome and augments dietary outcomes related to hepatic steatosis in mice. , 2020, Toxicology and applied pharmacology.

[22]  M. Bücking,et al.  Human biomonitoring of per- and polyfluoroalkyl substances in German blood plasma samples from 1982 to 2019. , 2020, Environment international.

[23]  Xiaojun Ge,et al.  Prevalence trends in non-alcoholic fatty liver disease at the global, regional and national levels, 1990–2017: a population-based observational study , 2020, BMJ Open.

[24]  E. Papadopoulou,et al.  Prenatal Exposure to Perfluoroalkyl Substances Associated With Increased Susceptibility to Liver Injury in Children , 2020, Hepatology.

[25]  G. Celano,et al.  Liver Steatosis, Gut-Liver Axis, Microbiome and Environmental Factors. A Never-Ending Bidirectional Cross-Talk , 2020, Journal of clinical medicine.

[26]  M. Longnecker,et al.  The concentration of several perfluoroalkyl acids in serum appears to be reduced by dietary fiber , 2020, medRxiv.

[27]  S. Kersten,et al.  Perfluorooctanoic acid (PFOA), perfluorooctane sulfonic acid (PFOS), and perfluorononanoic acid (PFNA) increase triglyceride levels and decrease cholesterogenic gene expression in human HepaRG liver cells , 2020, Archives of Toxicology.

[28]  R. Fry,et al.  Gut Microbiome Toxicity: Connecting the Environment and Gut Microbiome-Associated Diseases , 2020, Toxics.

[29]  B. Hennig,et al.  Healthful nutrition as a prevention and intervention paradigm to decrease the vulnerability to environmental toxicity or stressors and associated inflammatory disease risks. , 2020, Food frontiers.

[30]  B. Hennig,et al.  Prebiotic inulin consumption reduces dioxin-like PCB 126-mediated hepatotoxicity and gut dysbiosis in hyperlipidemic Ldlr deficient mice. , 2020, Environmental pollution.

[31]  K. Kleinman,et al.  Dietary characteristics associated with plasma concentrations of per- and polyfluoroalkyl substances among adults with pre-diabetes: Cross-sectional results from the Diabetes Prevention Program Trial. , 2020, Environment international.

[32]  H. Malhi,et al.  Pathogenesis of Nonalcoholic Steatohepatitis: An Overview , 2020, Hepatology communications.

[33]  J. Peters,et al.  Perfluorooctane Sulfonate Alters Gut Microbiota-Host Metabolic Homeostasis in Mice. , 2020, Toxicology.

[34]  Hongwen Sun,et al.  Distribution of novel and legacy per-/polyfluoroalkyl substances in serum and its associations with two glycemic biomarkers among Chinese adult men and women with normal blood glucose levels. , 2019, Environment international.

[35]  F. Javaudin,et al.  The effects of inulin on gut microbial composition: a systematic review of evidence from human studies , 2019, European Journal of Clinical Microbiology & Infectious Diseases.

[36]  Dean P. Jones,et al.  Perfluoroalkyl substances and severity of nonalcoholic fatty liver in Children: An untargeted metabolomics approach. , 2019, Environment international.

[37]  Jennifer Lu,et al.  Improved metagenomic analysis with Kraken 2 , 2019, Genome Biology.

[38]  Yuanxiang Jin,et al.  Bioaccumulation in the gut and liver causes gut barrier dysfunction and hepatic metabolism disorder in mice after exposure to low doses of OBS. , 2019, Environment international.

[39]  B. Stecher,et al.  Sequence and cultivation study of Muribaculaceae reveals novel species, host preference, and functional potential of this yet undescribed family , 2019, Microbiome.

[40]  B. Hennig,et al.  Hepatic metabolomics reveals that liver injury increases PCB 126-induced oxidative stress and metabolic dysfunction. , 2019, Chemosphere.

[41]  G. Shearer,et al.  Microbiota fermentation-NLRP3 axis shapes the impact of dietary fibres on intestinal inflammation , 2019, Gut.

[42]  Heather M. Wallace,et al.  Risk to human health related to the presence of perfluorooctane sulfonic acid and perfluorooctanoic acid in food , 2018, EFSA journal. European Food Safety Authority.

[43]  J. Allen,et al.  A Review of the Pathways of Human Exposure to Poly- and Perfluoroalkyl Substances (PFASs) and Present Understanding of Health Effects , 2018, Journal of Exposure Science & Environmental Epidemiology.

[44]  K. Lai,et al.  Dietary Exposure to the Environmental Chemical, PFOS on the Diversity of Gut Microbiota, Associated With the Development of Metabolic Syndrome , 2018, Front. Microbiol..

[45]  Jimmy D Bell,et al.  The effects of dietary supplementation with inulin and inulin‐propionate ester on hepatic steatosis in adults with non‐alcoholic fatty liver disease , 2018, Diabetes, obesity & metabolism.

[46]  Kassem M. Makki,et al.  The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. , 2018, Cell host & microbe.

[47]  H. Gilbert,et al.  Biochemistry of complex glycan depolymerisation by the human gut microbiota , 2018, FEMS microbiology reviews.

[48]  S. Summers,et al.  Could Ceramides Become the New Cholesterol? , 2018, Cell metabolism.

[49]  Curtis Huttenhower,et al.  bioBakery: a meta’omic analysis environment , 2017, Bioinform..

[50]  Y. Li,et al.  Half-lives of PFOS, PFHxS and PFOA after end of exposure to contaminated drinking water , 2017, Occupational and Environmental Medicine.

[51]  Diane Quagliani,et al.  Closing America’s Fiber Intake Gap , 2017, American journal of lifestyle medicine.

[52]  D. Ehresman,et al.  Organic Anion Transporting Polypeptides Contribute to the Disposition of Perfluoroalkyl Acids in Humans and Rats , 2016, Toxicological sciences : an official journal of the Society of Toxicology.

[53]  H. Tilg,et al.  Gut microbiome and liver diseases , 2016, Gut.

[54]  K. Lai,et al.  Fatty liver disease induced by perfluorooctane sulfonate: Novel insight from transcriptome analysis. , 2016, Chemosphere.

[55]  M. Blaut,et al.  Importance of propionate for the repression of hepatic lipogenesis and improvement of insulin sensitivity in high‐fat diet‐induced obesity , 2016, Molecular nutrition & food research.

[56]  D. van Sinderen,et al.  Bifidobacteria and Their Role as Members of the Human Gut Microbiota , 2016, Front. Microbiol..

[57]  Steven Salzberg,et al.  Bracken: Estimating species abundance in metagenomics data , 2016, bioRxiv.

[58]  W. Elkashef,et al.  Modified citrus pectin stops progression of liver fibrosis by inhibiting galectin-3 and inducing apoptosis of stellate cells. , 2016, Canadian journal of physiology and pharmacology.

[59]  W. Hinrichs,et al.  Inulin, a flexible oligosaccharide. II: Review of its pharmaceutical applications. , 2015, Carbohydrate polymers.

[60]  L. S. Haug,et al.  Examining confounding by diet in the association between perfluoroalkyl acids and serum cholesterol in pregnancy. , 2015, Environmental research.

[61]  S. Summers,et al.  Ceramides – Lipotoxic Inducers of Metabolic Disorders , 2015, Trends in Endocrinology & Metabolism.

[62]  S. Summers The ART of Lowering Ceramides. , 2015, Cell metabolism.

[63]  J. Forster,et al.  Na+/Taurocholate Cotransporting Polypeptide and Apical Sodium-Dependent Bile Acid Transporter Are Involved in the Disposition of Perfluoroalkyl Sulfonates in Humans and Rats. , 2015, Toxicological sciences : an official journal of the Society of Toxicology.

[64]  Yawei Wang,et al.  Differential accumulation and elimination behavior of perfluoroalkyl Acid isomers in occupational workers in a manufactory in China. , 2015, Environmental science & technology.

[65]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[66]  E. Botchwey,et al.  Acid sphingomyelinase is activated in sickle cell erythrocytes and contributes to inflammatory microparticle generation in SCD. , 2014, Blood.

[67]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[68]  G. Jiang,et al.  PFOS induced lipid metabolism disturbances in BALB/c mice through inhibition of low density lipoproteins excretion , 2014, Scientific Reports.

[69]  Brett J. Vanderford,et al.  Treatment of poly- and perfluoroalkyl substances in U.S. full-scale water treatment systems. , 2014, Water research.

[70]  Damià Barceló,et al.  Accumulation of perfluoroalkyl substances in human tissues. , 2013, Environment international.

[71]  Cole Trapnell,et al.  TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions , 2013, Genome Biology.

[72]  D. Ehresman,et al.  Comparative pharmacokinetics of perfluorooctanesulfonate (PFOS) in rats, mice, and monkeys. , 2012, Reproductive toxicology.

[73]  J. Giesy,et al.  PFOS-induced hepatic steatosis, the mechanistic actions on β-oxidation and lipid transport. , 2012, Biochimica et biophysica acta.

[74]  Guangchuang Yu,et al.  clusterProfiler: an R package for comparing biological themes among gene clusters. , 2012, Omics : a journal of integrative biology.

[75]  A. Ikari,et al.  Effects of dietary inulin, statin, and their co-treatment on hyperlipidemia, hepatic steatosis and changes in drug-metabolizing enzymes in rats fed a high-fat and high-sucrose diet , 2012, Nutrition & Metabolism.

[76]  W. Sanderson,et al.  Nutrition Can Modulate the Toxicity of Environmental Pollutants: Implications in Risk Assessment and Human Health , 2012, Environmental health perspectives.

[77]  G. Leonardi,et al.  Serum Perfluorooctanoate (PFOA) and Perfluorooctane Sulfonate (PFOS) Concentrations and Liver Function Biomarkers in a Population with Elevated PFOA Exposure , 2012, Environmental health perspectives.

[78]  J. Butenhoff,et al.  Multiplicity of nuclear receptor activation by PFOA and PFOS in primary human and rodent hepatocytes. , 2011, Toxicology.

[79]  M. V. van Erk,et al.  Perfluoroalkyl sulfonates cause alkyl chain length-dependent hepatic steatosis and hypolipidemia mainly by impairing lipoprotein production in APOE*3-Leiden CETP mice. , 2011, Toxicological sciences : an official journal of the Society of Toxicology.

[80]  C. Huttenhower,et al.  Metagenomic biomarker discovery and explanation , 2011, Genome Biology.

[81]  J. Depierre,et al.  Dietary exposure to perfluorooctanoate or perfluorooctane sulfonate induces hypertrophy in centrilobular hepatocytes and alters the hepatic immune status in mice. , 2010, International immunopharmacology.

[82]  A. Futerman,et al.  Mammalian ceramide synthases , 2010, IUBMB life.

[83]  Xianglin Shi,et al.  Perfluorooctane Sulfonate (PFOS) Induces Reactive Oxygen Species (ROS) Production in Human Microvascular Endothelial Cells: Role in Endothelial Permeability , 2010, Journal of toxicology and environmental health. Part A.

[84]  Efsa Panel on Dietetic Products Scientific Opinion on Dietary Reference Values for carbohydrates and dietary fibre , 2010 .

[85]  A. Voragen,et al.  Pectin, a versatile polysaccharide present in plant cell walls , 2009 .

[86]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[87]  Y. Hannun,et al.  The sphingolipid salvage pathway in ceramide metabolism and signaling. , 2008, Cellular signalling.

[88]  S. Summers,et al.  Sphingolipids, insulin resistance, and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism. , 2008, Endocrine reviews.

[89]  C. McClain,et al.  Using Nutrition for Intervention and Prevention against Environmental Chemical Toxicity and Associated Diseases , 2007, Environmental health perspectives.

[90]  J. Sugatani,et al.  Dietary Inulin Alleviates Hepatic Steatosis and Xenobiotics-Induced Liver Injury in Rats Fed a High-Fat and High-Sucrose Diet: Association with the Suppression of Hepatic Cytochrome P450 and Hepatocyte Nuclear Factor 4α Expression , 2006, Drug Metabolism and Disposition.

[91]  R. A. van den Berg,et al.  Centering, scaling, and transformations: improving the biological information content of metabolomics data , 2006, BMC Genomics.

[92]  R. Freedland,et al.  Effects of propionate on lipid biosynthesis in isolated rat hepatocytes. , 1990, The Journal of nutrition.

[93]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[94]  B. Abbott,et al.  Activation of Mouse and Human Peroxisome Proliferator-Activated Receptors ( , / , ) by Perfluorooctanoic Acid and Perfluorooctane Sulfonate , 2006 .

[95]  S. Summers,et al.  Ceramides in insulin resistance and lipotoxicity. , 2006, Progress in lipid research.

[96]  Susumu Goto,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 2000, Nucleic Acids Res..

[97]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .