Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 365
1.
Arch Toxicol ; 98(2): 409-424, 2024 Feb.
Article En | MEDLINE | ID: mdl-38099972

Arsenic, which can be divided into inorganic and organic arsenic, is a toxic metalloid that has been identified as a human carcinogen. A common source of arsenic exposure in seafood is arsenolipid, which is a complex structure of lipid-soluble organic arsenic compounds. At present, the known arsenolipid species mainly include arsenic-containing fatty acids (AsFAs), arsenic-containing hydrocarbons (AsHCs), arsenic glycophospholipids (AsPLs), and cationic trimethyl fatty alcohols (TMAsFOHs). Furthermore, the toxicity between different species is unique. However, the mechanism underlying arsenolipid toxicity and anabolism remain unclear, as arsenolipids exhibit a complex structure, are present at low quantities, and are difficult to extract and detect. Therefore, the objective of this overview is to summarize the latest research progress on methods to evaluate the toxicity and analyze the main speciation of arsenolipids in seafood. In addition, novel insights are provided to further elucidate the speciation, toxicity, and anabolism of arsenolipids and assess the risks on human health.


Arsenic , Arsenicals , Humans , Arsenic/toxicity , Fatty Acids/toxicity , Hydrocarbons/chemistry , Seafood/toxicity , Seafood/analysis
2.
Int J Toxicol ; 42(2_suppl): 5S-101S, 2023 08.
Article En | MEDLINE | ID: mdl-37279183

The Expert Panel for Cosmetic Ingredient Safety (Panel) assessed the safety of 274 polyglyceryl fatty acid esters. Each of the esters in this group is a polyether comprising 2 to 20 glyceryl residues, end-capped by esterification with simple carboxylic acids, such as fatty acids. Most of these ingredients are reported to function in cosmetics as skin-conditioning agents and/or surfactants. The Panel reviewed the available data and considered conclusions from their relevant previous reports, and determined that these ingredients are safe in cosmetics in the present practices of use and concentration described in this safety assessment when formulated to be non-irritating.


Consumer Product Safety , Cosmetics , Fatty Acids/toxicity , Cosmetics/toxicity , Esters/toxicity , Risk Assessment
3.
Arch Toxicol ; 97(5): 1247-1265, 2023 05.
Article En | MEDLINE | ID: mdl-36826474

3-Monochloropropane-1,2-diol (3-MCPD) is a chiral molecule naturally existing as a racemic mixture of (R)- and (S)-enantiomers. It was thoroughly investigated during the 1970s as a male antifertility drug until research was abandoned because of the side effects observed in toxicity studies. More than 20 years later, 3-MCPD, both in the free form and esterified to the fatty acids, was detected in vegetable oil and discovered to be a widespread contaminant in different processed foods. This review summarises the main toxicological studies on 3-MCPD and its esters. Current knowledge shows that the kidney and reproductive system are the primary targets of 3-MCPD toxicity, followed by neurological and immune systems. Despite uncertainties, in vivo studies suggest that renal and reproductive toxicity is mediated by toxic metabolites, leading to inhibition of glycolysis and energy depletion. Few acute, short-term, and subchronic toxicity studies have investigated the 3-MCPD esters. The pattern of toxicity was similar to that of free 3-MCPD. Some evidence suggests that the toxicity of 3-MCPD diesters may be milder than 3-MCPD, likely because of an incomplete enzymatic hydrolysis in the equivalent free form in the gastrointestinal tract. Further research to clarify absorption, metabolism, and long-term toxicity of 3-MCPD esters would be pivotal to improve the risk assessment of these compounds via food.


Esters , alpha-Chlorohydrin , Male , Humans , Esters/toxicity , Esters/metabolism , alpha-Chlorohydrin/toxicity , Fatty Acids/toxicity , Fatty Acids/metabolism , Hydrolysis , Kidney , Food Contamination/analysis
4.
J Biol Chem ; 299(3): 102937, 2023 03.
Article En | MEDLINE | ID: mdl-36690274

Defective autophagy and lipotoxicity are the hallmarks of nonalcoholic fatty liver disease. However, the precise molecular mechanism for the defective autophagy in lipotoxic conditions is not fully known. In the current study, we elucidated that activation of the mammalian target of rapamycin complex 1 (mTORC1)-G9a-H3K9me2 axis in fatty acid-induced lipotoxicity blocks autophagy by repressing key autophagy genes. The fatty acid-treated cells show mTORC1 activation, increased histone methyltransferase G9a levels, and suppressed autophagy as indicated by increased accumulation of the key autophagic cargo SQSTM1/p62 and decreased levels of autophagy-related proteins LC3II, Beclin1, and Atg7. Our chromatin immunoprecipitation analysis showed that decrease in autophagy was associated with increased levels of the G9a-mediated repressive H3K9me2 mark and decreased RNA polymerase II occupancy at the promoter regions of Beclin1 and Atg7 in fatty acid-treated cells. Inhibition of mTORC1 in fatty acid-treated cells decreased G9a-mediated H3K9me2 occupancy and increased polymerase II occupancy at Beclin1 and Atg7 promoters. Furthermore, mTORC1 inhibition increased the expression of Beclin1 and Atg7 in fatty acid-treated cells and decreased the accumulation of SQSTM1/p62. Interestingly, the pharmacological inhibition of G9a alone in fatty acid-treated cells decreased the H3K9me2 mark at Atg7 and Beclin1 promoters and restored the expression of Atg7 and Beclin1. Taken together, our findings have identified the mTORC1-G9a-H3K9me2 axis as a negative regulator of the autophagy pathway in hepatocellular lipotoxicity and suggest that the G9a-mediated epigenetic repression is mechanistically a key step during the repression of autophagy in lipotoxic conditions.


Autophagy , Fatty Acids , Histone Methyltransferases , Histones , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 1/metabolism , Histones/metabolism , Fatty Acids/toxicity , Autophagy/physiology , Epigenesis, Genetic , Histone Methyltransferases/metabolism , Hepatocytes/physiology , Hep G2 Cells , Gene Expression Regulation/drug effects , Palmitates/toxicity , Beclin-1/genetics , Beclin-1/metabolism , Promoter Regions, Genetic , Autophagosomes/genetics , Autophagosomes/metabolism , Humans
5.
FEBS Lett ; 596(11): 1424-1433, 2022 06.
Article En | MEDLINE | ID: mdl-35510803

Phosphatidylserine (PS) in the plasma membrane plays an important role in cell signaling and apoptosis. Cell degeneration is also linked to numerous amyloid diseases, pathologies that are associated with aggregation of misfolded proteins. In this work, we examine the effect of both saturated PS (DMPS) and unsaturated PS (DOPS and POPS) on the aggregation properties of insulin, as well as the structure and toxicity of insulin aggregates formed in the presence of these phospholipids. We found that the degree of unsaturation of fatty acids in PS alters the rate of insulin aggregation. We also found that toxicity of insulin-DMPS aggregates is significantly lower than the toxicity of DOPS- and POPS-insulin fibrils, whereas all these lipid-containing aggregates exert lower cell toxicity than insulin fibrils grown in a lipid-free environment.


Insulin , Phosphatidylserines , Amyloid/metabolism , Amyloidogenic Proteins , Fatty Acids/toxicity , Insulin/metabolism , Phosphatidylserines/chemistry , Phospholipids/metabolism
6.
Sci Total Environ ; 829: 154637, 2022 Jul 10.
Article En | MEDLINE | ID: mdl-35307418

The toxicity of pyrene (Pyr) and its chlorinated species have not be comprehensively and clearly elucidated. In this study, an integrated approach of metabolomics and transcriptomics were applied to evaluate the hepatotoxicity of Pyr and 1-chloropyrene (1-Cl-Pyr) at human exposure level, using human L02 hepatocytes. After 24 h exposure to Pyr and 1-Cl-Pyr at 5-500 nM, cell viability was not significantly changed. Transcriptomics results showed that exposure to Pyr and 1-Cl-Pyr at 5 and 50 nM obviously altered the gene expression profiles, but did not significantly induce the expression of genes strongly related to the activation of aryl hydrocarbon receptor (AhR), such as CYP1A1, CYP1B1, AHR, ARNT. Pyr and 1-Cl-Pyr both induced a notable metabolic perturbation to L02 cells. Glycerophospholipid metabolism was found to be the most significantly perturbed pathway after exposure to Pyr and 1-Cl-Pyr, indicating their potential damage to the cell membrane. The other significantly perturbed pathways were identified to be oxidative phosphorylation (OXPHOS), glycolysis, and fatty acid ß oxidation, all of which are related to energy production. Exposure to Pyr at 5 and 50 nM induced the up-regulation of fatty acid ß oxidation and OXPHOS. The similar result was observed after exposure to 5 nM 1-Cl-Pyr. In contrast, exposure to 50 nM 1-Cl-Pyr induced the down-regulation of OXPHOS by inhibiting the activity of complex I. The obtained results suggested that the modes of action of Pyr and 1-Cl-Pyr on energy production remarkably varied not only with molecular structure change but also with exposure concentration.


Chemical and Drug Induced Liver Injury , Polycyclic Aromatic Hydrocarbons , Cytochrome P-450 CYP1A1/metabolism , Fatty Acids/toxicity , Humans , Metabolomics , Polycyclic Aromatic Hydrocarbons/toxicity , Pyrenes/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Transcriptome
7.
Nutrients ; 13(7)2021 Jul 15.
Article En | MEDLINE | ID: mdl-34371939

A high-fat diet (HFD) and obesity are risk factors for many diseases including breast cancer. This is particularly important with close to 40% of the current adult population being overweight or obese. Previous studies have implicated that Mediterranean diets (MDs) partially protect against breast cancer. However, to date, the links between diet and breast cancer progression are not well defined. Therefore, to begin to define and assess this, we used an isocaloric control diet (CD) and two HFDs enriched with either olive oil (OOBD, high in oleate, and unsaturated fatty acid in MDs) or a milk fat-based diet (MFBD, high in palmitate and myristate, saturated fatty acids in Western diets) in a mammary polyomavirus middle T antigen mouse model (MMTV-PyMT) of breast cancer. Our data demonstrate that neither MFBD or OOBD altered the growth of primary tumors in the MMTV-PyMT mice. The examination of lung metastases revealed that OOBD mice exhibited fewer surface nodules and smaller metastases when compared to MFBD and CD mice. These data suggest that different fatty acids found in different sources of HFDs may alter breast cancer metastasis.


Breast Neoplasms/pathology , Diet, High-Fat/adverse effects , Dietary Fats/toxicity , Fatty Acids/toxicity , Lung Neoplasms/secondary , Milk/toxicity , Animal Feed , Animals , Antigens, Polyomavirus Transforming , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mammary Tumor Virus, Mouse/genetics , Olive Oil/toxicity , Risk Assessment , Risk Factors , Tumor Burden , Tumor Necrosis Factor-alpha/metabolism
8.
Reprod Toxicol ; 104: 58-67, 2021 09.
Article En | MEDLINE | ID: mdl-34246765

Perfluorononanoic acid (PFNA), a member of PFAS, is frequently detected in human blood and tissues, even in follicular fluid of women. The exposure of PFNA, but not PFOA and PFOS, is positively correlated with miscarriage and increased time to pregnancy. Toxicological studies indicated that PFNA exposure is associated with immunotoxicity, hepatotoxicity, developmental toxicity, and reproductive toxicity in animals. However, there is little information regarding the toxic effects of PFNA on oocyte maturation. In this study, we investigated the toxic effects of PFNA exposure on mouse oocyte maturation in vitro. Our results showed that 600 µM PFNA significantly inhibited germinal vesicle breakdown (GVBD) and polar body extrusion (PBE) in mouse oocytes. Our further study revealed that PFNA induced abnormal metaphase I (MI) spindle assembly, evidenced by malformed spindles and mislocalization of p-ERK1/2 in PFNA-treated oocytes. We also found that PFNA induced abnormal mitochondrial distribution and increased mitochondrial membrane potential. Consequently, PFNA increased reactive oxygen species (ROS) levels, leading to oxidative stress, DNA damage, and eventually early-stage apoptosis in oocytes. In addition, after 14 h culture, PFNA disrupted the formation of metaphase II (MII) spindle in most PFNA-treated oocytes with polar bodies. Collectively, our results indicate that PFNA interferes with oocyte maturation in vitro via disrupting spindle assembly, damaging mitochondrial functions, and inducing oxidative stress, DNA damage, and early-stage apoptosis.


Fatty Acids/toxicity , Fluorocarbons/toxicity , Mitochondria/drug effects , Oxidative Stress/physiology , Animals , Female , Metaphase , Mice , Oocytes/drug effects , Oogenesis , Oxidative Stress/drug effects
9.
Environ Health ; 20(1): 76, 2021 06 30.
Article En | MEDLINE | ID: mdl-34193151

BACKGROUND: Per- and polyfluoroalkyl substances (PFAS) and polybrominated diphenyl ethers (PBDEs) are endocrine disrupting chemicals with widespread exposures across the U.S. given their abundance in consumer products. PFAS and PBDEs are associated with reproductive toxicity and adverse health outcomes, including certain cancers. PFAS and PBDEs may affect health through alternations in telomere length. In this study, we examined joint associations between prenatal exposure to PFAS, PBDEs, and maternal and newborn telomere length using mixture analyses, to characterize effects of cumulative environmental chemical exposures. METHODS: Study participants were enrolled in the Chemicals in Our Bodies (CIOB) study, a demographically diverse cohort of pregnant people and children in San Francisco, CA. Seven PFAS (ng/mL) and four PBDEs (ng/g lipid) were measured in second trimester maternal serum samples. Telomere length (T/S ratio) was measured in delivery cord blood of 292 newborns and 110 second trimester maternal whole blood samples. Quantile g-computation was used to assess the joint associations between groups of PFAS and PBDEs and newborn and maternal telomere length. Groups considered were: (1) all PFAS and PBDEs combined, (2) PFAS, and (3) PBDEs. Maternal and newborn telomere length were modeled as separate outcomes. RESULTS: T/S ratios in newborn cord and maternal whole blood were moderately correlated (Spearman ρ = 0.31). In mixtures analyses, a simultaneous one quartile increase in all PFAS and PBDEs was associated with a small increase in newborn (mean change per quartile increase = 0.03, 95% confidence interval [CI] = -0.03, 0.08) and maternal telomere length (mean change per quartile increase = 0.03 (95% CI = -0.03, 0.09). When restricted to maternal-fetal paired samples (N = 76), increasing all PFAS and PBDEs combined was associated with a strong, positive increase in newborn telomere length (mean change per quartile increase = 0.16, 95% CI = 0.03, 0.28). These associations were primarily driven by PFAS (mean change per quartile increase = 0.11 [95% CI = 0.01, 0.22]). No associations were observed with maternal telomere length among paired samples. CONCLUSIONS: Our findings suggest that PFAS and PBDEs may be positively associated with newborn telomere length.


Environmental Pollutants/toxicity , Flame Retardants/toxicity , Fluorocarbons/toxicity , Halogenated Diphenyl Ethers/toxicity , Prenatal Exposure Delayed Effects , Telomere/drug effects , Adult , Biological Monitoring , Environmental Pollutants/analysis , Fatty Acids/analysis , Fatty Acids/toxicity , Female , Flame Retardants/analysis , Fluorocarbons/analysis , Halogenated Diphenyl Ethers/analysis , Humans , Infant, Newborn , Male , Maternal Exposure , Maternal-Fetal Exchange , Pregnancy , Sulfonic Acids/analysis , Sulfonic Acids/toxicity
10.
Neurochem Int ; 148: 105050, 2021 09.
Article En | MEDLINE | ID: mdl-33945834

Neurons spurn hydrogen-rich fatty acids for energizing oxidative ATP synthesis, contrary to other cells. This feature has been mainly attributed to a lower yield of ATP per reduced oxygen, as compared to glucose. Moreover, the use of fatty acids as hydrogen donor is accompanied by severe ß-oxidation-associated ROS generation. Neurons are especially susceptible to detrimental activities of ROS due to their poor antioxidative equipment. It is also important to note that free fatty acids (FFA) initiate multiple harmful activities inside the cells, particularly on phosphorylating mitochondria. Several processes enhance FFA-linked lipotoxicity in the cerebral tissue. Thus, an uptake of FFA from the circulation into the brain tissue takes place during an imbalance between energy intake and energy expenditure in the body, a situation similar to that during metabolic syndrome and fat-rich diet. Traumatic or hypoxic brain injuries increase hydrolytic degradation of membrane phospholipids and, thereby elevate the level of FFA in neural cells. Accumulation of FFA in brain tissue is markedly associated with some inherited neurological disorders, such as Refsum disease or X-linked adrenoleukodystrophy (X-ALD). What are strategies protecting neurons against FFA-linked lipotoxicity? Firstly, spurning the ß-oxidation pathway in mitochondria of neurons. Secondly, based on a tight metabolic communication between neurons and astrocytes, astrocytes donate metabolites to neurons for synthesis of antioxidants. Further, neuronal autophagy of ROS-emitting mitochondria combined with the transfer of degradation-committed FFA for their disposal in astrocytes, is a potent protective strategy against ROS and harmful activities of FFA. Finally, estrogens and neurosteroids are protective as triggers of ERK and PKB signaling pathways, consequently initiating the expression of various neuronal survival genes via the formation of cAMP response element-binding protein (CREB).


Brain/metabolism , Fatty Acids/toxicity , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Brain/drug effects , Energy Metabolism/physiology , Fatty Acids/metabolism , Humans , Mitochondria/drug effects , Neurons/metabolism , Oxidative Stress/physiology
11.
J Cell Physiol ; 236(10): 7033-7044, 2021 10.
Article En | MEDLINE | ID: mdl-33738797

Fatty acids (FA) exert physiological and pathophysiological effects leading to changes in skeletal muscle metabolism and function, however, in vitro models to investigate these changes are limited. These experiments sought to establish the effects of physiological and pathophysiological concentrations of exogenous FA upon the function of tissue engineered skeletal muscle (TESkM). Cultured initially for 14 days, C2C12 TESkM was exposed to FA-free bovine serum albumin alone or conjugated to a FA mixture (oleic, palmitic, linoleic, and α-linoleic acids [OPLA] [ratio 45:30:24:1%]) at different concentrations (200 or 800 µM) for an additional 4 days. Subsequently, TESkM morphology, functional capacity, gene expression and insulin signaling were analyzed. There was a dose response increase in the number and size of lipid droplets within the TESkM (p < .05). Exposure to exogenous FA increased the messenger RNA expression of genes involved in lipid storage (perilipin 2 [p < .05]) and metabolism (pyruvate dehydrogenase lipoamide kinase isozyme 4 [p < .01]) in a dose dependent manner. TESkM force production was reduced (tetanic and single twitch) (p < .05) and increases in transcription of type I slow twitch fiber isoform, myosin heavy chain 7, were observed when cultured with 200 µM OPLA compared to control (p < .01). Four days of OPLA exposure results in lipid accumulation in TESkM which in turn results in changes in muscle function and metabolism; thus, providing insight ito the functional and mechanistic changes of TESkM in response to exogenous FA.


Fatty Acids/toxicity , Lipid Droplets/drug effects , Lipid Metabolism/drug effects , Muscle, Skeletal/drug effects , Myoblasts, Skeletal/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation , Insulin/pharmacology , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Mice , Muscle Strength/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Tissue Engineering
12.
Toxicol Appl Pharmacol ; 415: 115440, 2021 03 15.
Article En | MEDLINE | ID: mdl-33549592

Perfluoroundecanoic acid (PFUnA) is one of long-chain perfluoroalkyl carboxylic acids. However, the effect of PFUnA on pubertal development of Leydig cells remains unclear. The goal of this study was to investigate the effect of PFUnA on Leydig cell development in pubertal male rats. We orally dosed male Sprague-Dawley rats (age 35 days) with PFUnA at doses of 0, 1, 5, and 10 mg/kg/day from postnatal day (PND) 35 to PND 56. Serum testosterone and luteinizing hormone levels were remarkably reduced by PFUnA at ≥1 mg/kg while serum follicle-stimulating hormone levels were lowered at 5 and 10 mg/kg. PFUnA down-regulated the expression of Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Hsd11b1, Insl3, Nr5a1, Fshr, Dhh, Sod1, and Sod2 and their proteins in the testis and the expression of Lhb and Fshb in the pituitary. PFUnA reduced Leydig cell number at 5 and 10 mg/kg. PFUnA induced oxidative stress and increased autophagy. These may result from the inhibition of phosphorylation of mTOR, AKT1, AKT2, and ERK1/2 in the testis. In conclusion, PFUnA exhibits inhibitory effects on pubertal Leydig cell development possibly via inducing oxidative stress and increasing autophagy.


Autophagy/drug effects , Fatty Acids/toxicity , Fluorocarbons/toxicity , Leydig Cells/drug effects , Oxidative Stress/drug effects , Age Factors , Animals , Autophagy-Related Proteins/metabolism , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Enzymologic , Leydig Cells/metabolism , Leydig Cells/pathology , Luteinizing Hormone/blood , Male , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats, Sprague-Dawley , Sexual Development , Signal Transduction , Sperm Count , Spermatozoa/drug effects , Spermatozoa/pathology , Testosterone/blood
13.
Sci Rep ; 11(1): 1020, 2021 01 13.
Article En | MEDLINE | ID: mdl-33441911

Stressful conditions during development can have sub-lethal consequences on organisms aside from mortality. Using previously reported in-hive residues from commercial colonies, we examined how multi-pesticide exposure can influence honey bee (Apis mellifera) queen health. We reared queens in beeswax cups with or without a pesticide treatment within colonies exposed to treated or untreated pollen supplement. Following rearing, queens were open-mated and then placed into standard hive equipment in an "artificial swarm" to measure subsequent colony growth. Our treated wax had a pesticide Hazard Quotient comparable to the average in beeswax from commercial colonies, and it had no measurable effects on queen phenotype. Conversely, colonies exposed to pesticide-treated pollen had a reduced capacity for viable queen production, and among surviving queens from these colonies we observed lower sperm viability. We found no difference in queen mating number across treatments. Moreover, we measured lower brood viability in colonies later established by queens reared in treated-pollen colonies. Interestingly, royal jelly from colonies exposed to treated pollen contained negligible pesticide residues, suggesting the indirect social consequences of colony-level pesticide exposure on queen quality. These findings highlight how conditions during developmental can impact queens long into adulthood, and that colony-level pesticide exposure may do so indirectly.


Bees/drug effects , Bees/physiology , Pesticides/toxicity , Animals , Bees/growth & development , Fatty Acids/chemistry , Fatty Acids/toxicity , Female , Male , Oviposition/drug effects , Pesticide Residues/analysis , Pesticide Residues/toxicity , Pesticides/analysis , Phenotype , Pollen/chemistry , Pollen/toxicity , Reproduction/drug effects , Social Behavior , Sperm Count , Waxes/chemistry , Waxes/toxicity
14.
Arch Toxicol ; 95(1): 355-374, 2021 01.
Article En | MEDLINE | ID: mdl-32909075

Nuclear receptors (NRs) are key regulators of energy homeostasis, body development, and sexual reproduction. Xenobiotics binding to NRs may disrupt natural hormonal systems and induce undesired adverse effects in the body. However, many chemicals of concerns have limited or no experimental data on their potential or lack-of-potential endocrine-disrupting effects. Here, we propose a virtual screening method based on molecular docking for predicting potential endocrine-disrupting chemicals (EDCs) that bind to NRs. For 12 NRs, we systematically analyzed how multiple crystal structures can be used to distinguish actives and inactives found in previous high-throughput experiments. Our method is based on (i) consensus docking scores from multiple structures at a single functional state (agonist-bound or antagonist-bound), (ii) multiple functional states (agonist-bound and antagonist-bound), and (iii) multiple pockets (orthosteric site and alternative sites) of these NRs. We found that the consensus enrichment from multiple structures is better than or comparable to the best enrichment from a single structure. The discriminating power of this consensus strategy was further enhanced by a chemical similarity-weighted scoring scheme, yielding better or comparable enrichment for all studied NRs. Applying this optimized method, we screened 252 fatty acids against peroxisome proliferator-activated receptor gamma (PPARγ) and successfully identified 3 previously unknown fatty acids with Kd = 100-250 µM including two furan fatty acids: furannonanoic acid (FNA) and furanundecanoic acid (FUA), and one cyclopropane fatty acid: phytomonic acid (PTA). These results suggested that the proposed method can be used to rapidly screen and prioritize potential EDCs for further experimental evaluations.


Endocrine Disruptors/metabolism , Fatty Acids/metabolism , Molecular Docking Simulation , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Toxicity Tests , Binding Sites , Databases, Protein , Endocrine Disruptors/chemistry , Endocrine Disruptors/toxicity , Fatty Acids/chemistry , Fatty Acids/toxicity , Feasibility Studies , Ligands , PPAR gamma/chemistry , PPAR gamma/drug effects , Protein Binding , Protein Conformation , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/drug effects , Risk Assessment , Structure-Activity Relationship , Surface Plasmon Resonance
15.
Mol Cell Endocrinol ; 520: 111076, 2021 01 15.
Article En | MEDLINE | ID: mdl-33159991

Calcium/calmodulin-dependent serine protein kinase (CASK) knockdown reduces insulin vesicle docking to cell membranes. Here, we explored CASK interactions with other proteins during insulin secretion. Using co-immunoprecipitation, liquid chromatography-mass spectrometry and bioinformatic analysis, we identified that CASK, Adapter protein X11 alpha (APBA1), and Syntaxin binding protein 1 (STXBP1) formed tripartite complex during insulin secretion. CASK enhanced APBA1-STXBP1 interaction and mediated their traffic from cytoplasm to plasma membrane during insulin release. High fatty acid stimulation decreased insulin secretion along with CASK, APBA1, and STXBP1 expression; Cask overexpression enhanced CASK/APBA1/STXBP1 tripartite complex function, and may thereby rescue lipotoxicity-induced insulin-release defects. Collectively, our results illustrated the function of CASK in insulin granules exocytosis, which broadens the underlying mechanism of insulin secretion and highlights the clinical potential of CASK as a drug target of type 2 Diabetes Mellitus (T2DM).


Adaptor Proteins, Signal Transducing/metabolism , Guanylate Kinases/metabolism , Insulin Secretion , Membrane Proteins/metabolism , Munc18 Proteins/metabolism , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Exocytosis/drug effects , Fatty Acids/toxicity , Gene Ontology , Insulin Secretion/drug effects , Male , Mice, Inbred C57BL , Protein Binding/drug effects , Rats
16.
Chemosphere ; 262: 128012, 2021 Jan.
Article En | MEDLINE | ID: mdl-33182161

Due to global restriction on perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA), the use of long-chain perfluoroalkyl substances (PFASs, C > 8) and their environmental occurrences have increased. PFOS and PFOA have been known for thyroid disruption, however, knowledge is still limited on thyroid disrupting effects of long-chain PFASs (C > 10). In this study, two long-chain perfluorinated carboxylic acids (PFCAs), i.e., perfluoroundecanoic acid (PFUnDA) and perfluorotridecanoic acid (PFTrDA), were chosen and investigated for thyroid disrupting effects, using zebrafish embryo/larvae and rat pituitary cell line (GH3). For comparison, PFOA was also added as a test chemical and also investigated for its thyroid disruption potential. Following a 5 d exposure to PFTrDA, zebrafish larvae showed upregulation of the genes responsible for thyroid hormone synthesis (tshß, nkx2.1, nis, tpo, mct8) and (de)activation (dio1, dio2). In contrast, both PFUnDA and PFOA induced no regulatory changes except for upregulation of a thyroid metabolism related gene (ugt1ab). Morphological changes such as decreased eyeball size, increased yolk sac size, or deflated swim bladder, occurred following exposure to PFUnDA, PFTrDA, and PFOA. In GH3 cells, exposure to PFUnDA and PFTrDA upregulated Tshß gene, suggesting that these PFCAs increase thyroid hormone synthesis through stimulation by Tsh. In summary, both long-chain PFCAs could cause transcriptional changes of thyroid regulating genes that may lead to increased malformation of the zebrafish larvae, but the pathway of thyroid disruption appears to be different by the chain length. Confirmation and validation in adult fish following long term exposure are warranted.


Decanoic Acids/toxicity , Endocrine Disruptors/toxicity , Fatty Acids/toxicity , Fluorocarbons/toxicity , Pituitary Gland/drug effects , Thyroid Gland/drug effects , Thyroid Hormones/metabolism , Zebrafish/metabolism , Animals , Cell Line , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Gene Expression/drug effects , Larva/drug effects , Larva/metabolism , Pituitary Gland/metabolism , Rats , Thyroid Gland/metabolism , Thyroid Hormones/genetics
17.
J Appl Toxicol ; 41(7): 1148-1162, 2021 07.
Article En | MEDLINE | ID: mdl-33145810

Cetylated fatty acids (CFAs) are a group of fats that contain a single ester group within a hydrocarbon chain, which are reported to have beneficial biological effects. A novel mixture of CFAs produced by combining fatty acids derived from refined olive oil with cetyl esters (Lipocet®) is proposed for use as a food ingredient and was therefore subjected to a nonclinical safety assessment. The safety of Lipocet® was evaluated in a bacterial reverse mutation test and an in vitro mammalian cell micronucleus test, followed by a 90-day oral (gavage) toxicity study. In the 90-day study, Sprague-Dawley rats were administered with the vehicle (corn oil) or Lipocet® at 1,500, 3,000, or 4,500 mg/kg body weight/day for 90 days. A comparator reference control group received noncetylated fatty acids derived from olive oil at 4,500 mg/kg body weight/day to identify any effects that may be expected following consumption of high doses of fat. Lipocet® was nongenotoxic in vitro. In the 90-day study, changes observed in hematological and clinical biochemistry parameters were minor in nature and/or showed poor dose dependency. Histopathology findings in the gastrointestinal tract and lungs were noted but were not associated with a clear dose response and were likely incidental. Moreover, Lipocet® was just as well tolerated as the reference control. Therefore, 4,500 mg/kg body weight/day (the highest dose tested) was considered the no-observed-adverse-effect-level. These results support the safety of Lipocet® for use as a food ingredient.


Fatty Acids/toxicity , Animals , Carcinogens/administration & dosage , DNA Damage , Male , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Toxicity Tests, Subchronic
18.
Int J Toxicol ; 39(3_suppl): 93S-126S, 2020.
Article En | MEDLINE | ID: mdl-33203266

The Expert Panel for Cosmetic Ingredient Safety (Panel) assessed the safety of 44 monoglyceryl monoesters that are structurally constituted as the esterification products of glycerin and carboxylic acids (the majority of which are fatty acids); 36 of these monoesters were previously reviewed by the Panel, and 8 are reviewed herein for the first time. Most of the monoglyceryl monoesters have several reported functions in cosmetics, but the most common function among the ingredients is skin conditioning agent; a few are reported to function only as surfactant-emulsifying agents. The Panel reviewed relevant new data, including frequency and concentration of use and considered the data from previous Cosmetic Ingredient Review reports. The Panel concluded that these ingredients are safe in cosmetics in the present practices of use and concentration described in this safety assessment.


Cosmetics , Fatty Acids , Monoglycerides , Animals , Consumer Product Safety , Cosmetics/adverse effects , Cosmetics/chemistry , Cosmetics/toxicity , Fatty Acids/adverse effects , Fatty Acids/chemistry , Fatty Acids/toxicity , Humans , Mice , Monoglycerides/adverse effects , Monoglycerides/chemistry , Monoglycerides/toxicity , Rats , Toxicity Tests
19.
Sci Rep ; 10(1): 13523, 2020 08 11.
Article En | MEDLINE | ID: mdl-32782332

Autophagy, an integral part of the waste recycling process, plays an important role in cellular physiology and pathophysiology. Impaired autophagic flux causes ectopic lipid deposition, which is defined as the accumulation of lipids in non-adipose tissue. Ectopic lipid accumulation is observed in patients with cardiometabolic syndrome, including obesity, diabetes, insulin resistance, and cardiovascular complications. Metformin is the first line of treatment for type 2 diabetes, and one of the underlying mechanisms for the anti-diabetic effect of metformin is mediated by the stimulation of AMP-activated protein kinase (AMPK). Because the activation of AMPK is crucial for the initiation of autophagy, we hypothesize that metformin reduces the accumulation of lipid droplets by increasing autophagic flux in vascular endothelial cells. Incubation of vascular endothelial cells with saturated fatty acid (SFA) increased the accumulation of lipid droplets and impaired autophagic flux. We observed that the accumulation of lipid droplets was reduced, and the autophagic flux was enhanced by treatment with metformin. The knock-down of AMPKα by using siRNA blunted the effect of metformin. Furthermore, treatment with SFA or inhibition of autophagy increased leukocyte adhesion, whereas treatment with metformin decreased the SFA-induced leukocyte adhesion. The results suggest a novel mechanism by which metformin protects vascular endothelium from SFA-induced ectopic lipid accumulation and pro-inflammatory responses. In conclusion, improving autophagic flux may be a therapeutic strategy to protect endothelial function from dyslipidemia and diabetic complications.


Autophagy , Carnitine O-Palmitoyltransferase/physiology , Endothelium, Vascular/drug effects , Fatty Acids/toxicity , Hypoglycemic Agents/pharmacology , Inflammation/drug therapy , Metformin/pharmacology , AMP-Activated Protein Kinases , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
J Biol Chem ; 295(25): 8628-8635, 2020 06 19.
Article En | MEDLINE | ID: mdl-32393576

Excess fatty acid accumulation in nonadipose tissues leads to cell dysfunction and cell death that is linked to the pathogenesis of inherited and acquired human diseases. Study of this process, known as lipotoxicity, has provided new insights into the regulation of lipid homeostasis and has revealed new molecular pathways involved in lipid-induced cellular stress. The discovery that disruption of specific small nucleolar RNAs protects against fatty acid-induced cell death and remodels metabolism in vivo opens new opportunities for understanding how nutrient signals influence cellular and systemic metabolic homeostasis through RNA biology.


Cell Death , Fatty Acids/metabolism , RNA, Small Nucleolar/metabolism , Animals , Cell Death/drug effects , Endoplasmic Reticulum Stress/drug effects , Fatty Acids/toxicity , Humans , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Reactive Oxygen Species/metabolism
...