In vivo characterisation of the toxicological properties of DPhP, one of the main degradation products of aryl phosphate esters

Background Aryl phosphate esters (APEs), a main class of organophosphorus ester molecules, are widely used and commonly present in the environment. Health hazards associated with these compounds remain largely unknown and the effects of diphenyl phosphate (DPhP), one of their most frequent derivatives in human samples, are poorly characterised. Objective Our aim was to investigate whether DPhP per se may represent a more relevant marker of exposure to APEs and determine its potential deleterious biological effects in chronically exposed mice. Methods Conventional animals (FVB mice) were acutely (intravenous or oral gavage) or chronically (0.1 mg.mL-1, 1 mg.mL-1, 10 mg.mL-1 in drink water) exposed to relevant doses of DPhP or triphenyl phosphate (TPhP), one of its main precursors in the environment. Both molecules were measured in blood and other relevant tissues by liquid chromatography-mass spectrometry (LC-MS). Biological effects of chronic DPhP exposure were addressed through liver multi-omics analysis combining mRNA extraction and sequencing to high resolution LC-MS to determine the corresponding metabolic profile. Deep statistical exploration was performed to extract correlated information, guiding further physiological analyses (immunohistochemistry (IHC) and animal growth measurement). Results Acute and chronic exposure to DPhP led to significant levels of this molecule in blood and other tissues, an effect missing with TPhP. Multi-omics analysis confirmed the existence of biological effects of DPhP, even at a very low dose of 0.1 mg.mL-1 in drinking water. Chemical structural homology and pathway mapping demonstrated a clear reduction of the fatty-acid catabolic processes centred on acylcarnitine and mitochondrial β-oxidation. Interestingly, mRNA expression confirmed and extended these observations by demonstrating at all tested doses the overall repression of genes involved in lipid catabolic processes and regulated by PPARα, a master regulator of β-oxidation and its associated ketogenesis. IHC analysis confirmed the alteration of these pathways by showing a specific downregulation of Hmgcs2, a kernel target gene of PPARα, at all doses tested, and surprisingly, a strong reduction of the lipid droplet content only at the highest dose. Overall, DPhP absorption led to weight loss, which was significant using the highest dose. Conclusions Our results suggest that in mice, the effects of chronic exposure to DPhP, even at a low dose, are not negligible. Fatty acid metabolism in the liver in particular is essential for controlling fast and feast periods with adverse consequences on the overall physiology. Therefore, the impact of DPhP on circulating fat, cardiovascular and metabolic disease incidence deserves, in light of our results, further investigations.

[1]  P. Roman,et al.  Microbiota and organophosphates. , 2019, Neurotoxicology.

[2]  Yong Yuan,et al.  Enhanced degradation of triphenyl phosphate (TPHP) in bioelectrochemical systems: Kinetics, pathway and degradation mechanisms. , 2019, Environmental pollution.

[3]  K. Bock Aryl hydrocarbon receptor (AHR): From selected human target genes and crosstalk with transcription factors to multiple AHR functions. , 2019, Biochemical pharmacology.

[4]  R. Deberardinis,et al.  MYC promotes tryptophan uptake and metabolism by the kynurenine pathway in colon cancer , 2019, Genes & development.

[5]  S. Harrad,et al.  First insight into human extrahepatic metabolism of flame retardants: Biotransformation of EH-TBB and Firemaster-550 components by human skin subcellular fractions. , 2019, Chemosphere.

[6]  N. Yajima,et al.  Kynurenine signaling through the aryl hydrocarbon receptor maintains the undifferentiated state of human embryonic stem cells , 2019, Science Signaling.

[7]  K. Bock Aryl hydrocarbon receptor (AHR) functions in NAD+ metabolism, myelopoiesis and obesity , 2019, Biochemical pharmacology.

[8]  R. Letcher,et al.  A review on organophosphate Ester (OPE) flame retardants and plasticizers in foodstuffs: Levels, distribution, human dietary exposure, and future directions. , 2019, Environment international.

[9]  H. Stapleton,et al.  Diphenyl Phosphate-Induced Toxicity During Embryonic Development. , 2019, Environmental science & technology.

[10]  R. Halden,et al.  A nationwide survey of 31 organophosphate esters in sewage sludge from the United States. , 2019, The Science of the total environment.

[11]  Jianying Hu,et al.  Urinary biomarkers for assessment of human exposure to monomeric aryl phosphate flame retardants. , 2019, Environment international.

[12]  K. Kannan,et al.  Concentrations and Dietary Exposure to Organophosphate Esters in Foodstuffs from Albany, New York, United States. , 2018, Journal of agricultural and food chemistry.

[13]  Roya Jamarani,et al.  How Green is Your Plasticizer? , 2018, Polymers.

[14]  Miaomiao Teng,et al.  Neonatal triphenyl phosphate and its metabolite diphenyl phosphate exposure induce sex- and dose-dependent metabolic disruptions in adult mice. , 2018, Environmental pollution.

[15]  A. Ballesteros-Gómez,et al.  Presence of diphenyl phosphate and aryl-phosphate flame retardants in indoor dust from different microenvironments in Spain and the Netherlands and estimation of human exposure. , 2018, Environment international.

[16]  Lixi Zeng,et al.  Organophosphate Triesters and Diester Degradation Products in Municipal Sludge from Wastewater Treatment Plants in China: Spatial Patterns and Ecological Implications. , 2017, Environmental science & technology.

[17]  O. Fiehn,et al.  Chemical Similarity Enrichment Analysis (ChemRICH) as alternative to biochemical pathway mapping for metabolomic datasets , 2017, Scientific Reports.

[18]  E. Atlas,et al.  Firemaster® 550 and its components isopropylated triphenyl phosphate and triphenyl phosphate enhance adipogenesis and transcriptional activity of peroxisome proliferator activated receptor (Pparγ) on the adipocyte protein 2 (aP2) promoter , 2017, PloS one.

[19]  J. Eeckhoute,et al.  Distinct but complementary contributions of PPAR isotypes to energy homeostasis. , 2017, The Journal of clinical investigation.

[20]  N. Van den Eede,et al.  Does Biotransformation of Aryl Phosphate Flame Retardants in Blood Cast a New Perspective on Their Debated Biomarkers? , 2016, Environmental science & technology.

[21]  R. Letcher,et al.  Organophosphate Flame Retardants and Plasticizers in Aqueous Solution: pH-Dependent Hydrolysis, Kinetics, and Pathways. , 2016, Environmental science & technology.

[22]  R. Letcher,et al.  Determination of glucuronide conjugates of hydroxyl triphenyl phosphate (OH-TPHP) metabolites in human urine and its use as a biomarker of TPHP exposure. , 2016, Chemosphere.

[23]  H. Stapleton,et al.  Nail polish as a source of exposure to triphenyl phosphate. , 2016, Environment international.

[24]  S. Kersten,et al.  The impact of PPARα activation on whole genome gene expression in human precision cut liver slices , 2015, BMC Genomics.

[25]  Yuanxiang Jin,et al.  Exposure of male mice to two kinds of organophosphate flame retardants (OPFRs) induced oxidative stress and endocrine disruption. , 2015, Environmental toxicology and pharmacology.

[26]  N. Van den Eede,et al.  In vitro human metabolism of the flame retardant resorcinol bis(diphenylphosphate) (RDP). , 2015, Environmental science & technology.

[27]  Adrian Covaci,et al.  In vitro metabolism of 2-ethylhexyldiphenyl phosphate (EHDPHP) by human liver microsomes. , 2015, Toxicology letters.

[28]  S. Kennedy,et al.  Rapid in vitro metabolism of the flame retardant triphenyl phosphate and effects on cytotoxicity and mRNA expression in chicken embryonic hepatocytes. , 2014, Environmental science & technology.

[29]  H. Patisaul,et al.  In vitro assessment of human nuclear hormone receptor activity and cytotoxicity of the flame retardant mixture FM 550 and its triarylphosphate and brominated components. , 2014, Toxicology letters.

[30]  Takahiko Yoshida,et al.  In vitro endocrine disruption potential of organophosphate flame retardants via human nuclear receptors. , 2013, Toxicology.

[31]  J. Meeker,et al.  Urinary Metabolites of Organophosphate Flame Retardants: Temporal Variability and Correlations with House Dust Concentrations , 2013, Environmental health perspectives.

[32]  P. Schirmacher,et al.  Adipophilin/perilipin‐2 as a lipid droplet‐specific marker for metabolically active cells and diseases associated with metabolic dysregulation , 2013, Histopathology.

[33]  Oliver Fiehn,et al.  MetaMapp: mapping and visualizing metabolomic data by integrating information from biochemical pathways and chemical and mass spectral similarity , 2012, BMC Bioinformatics.

[34]  W. Wahli,et al.  Proline- and acidic amino acid-rich basic leucine zipper proteins modulate peroxisome proliferator-activated receptor α (PPARα) activity , 2011, Proceedings of the National Academy of Sciences.

[35]  John D. Meeker,et al.  House Dust Concentrations of Organophosphate Flame Retardants in Relation to Hormone Levels and Semen Quality Parameters , 2009, Environmental health perspectives.

[36]  H. Waterham,et al.  Novel metabolic and molecular findings in hepatic carnitine palmitoyltransferase I deficiency. , 2005, Molecular genetics and metabolism.

[37]  G. Watts,et al.  Differential regulation of lipoprotein kinetics by atorvastatin and fenofibrate in subjects with the metabolic syndrome. , 2003, Diabetes.

[38]  S. Dey,et al.  A novel pathway of prostacyclin signaling-hanging out with nuclear receptors. , 2002, Endocrinology.

[39]  J Auwerx,et al.  PPARalpha and PPARgamma activators direct a distinct tissue‐specific transcriptional response via a PPRE in the lipoprotein lipase gene. , 1996, The EMBO journal.

[40]  C. Cerniglia,et al.  Fungal metabolism of tert-butylphenyl diphenyl phosphate , 1985, Applied and environmental microbiology.

[41]  M. Takeda,et al.  Metabolism of phosphoric acid triesters by rat liver homogenate , 1984, Bulletin of environmental contamination and toxicology.