Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.089
Filter
1.
J Biol Chem ; 297(4): 101147, 2021 10.
Article in English | MEDLINE | ID: mdl-34520756

ABSTRACT

The heterogeneity of respirable particulates and compounds complicates our understanding of transcriptional responses to air pollution. Here, we address this by applying precision nuclear run-on sequencing and the assay for transposase-accessible chromatin sequencing to measure nascent transcription and chromatin accessibility in airway epithelial cells after wood smoke particle (WSP) exposure. We used transcription factor enrichment analysis to identify temporally distinct roles for ternary response factor-serum response factor complexes, the aryl hydrocarbon receptor (AHR), and NFκB in regulating transcriptional changes induced by WSP. Transcription of canonical targets of the AHR, such as CYP1A1 and AHRR, was robustly increased after just 30 min of WSP exposure, and we discovered novel AHR-regulated pathways and targets including the DNA methyltransferase, DNMT3L. Transcription of these genes and associated enhancers rapidly returned to near baseline by 120 min after exposure. The kinetics of AHR- and NFκB-regulated responses to WSP were distinguishable based on the timing of both transcriptional responses and chromatin remodeling, with induction of several cytokines implicated in maintaining NFκB-mediated responses through 120 min of exposure. In aggregate, our data establish a direct and primary role for AHR in mediating airway epithelial responses to WSP and identify crosstalk between AHR and NFκB signaling in controlling proinflammatory gene expression. This work also defines an integrated genomics-based strategy for deconvoluting multiplexed transcriptional responses to heterogeneous environmental exposures.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Repressor Proteins/metabolism , Signal Transduction , Smoke/adverse effects , Transcription, Genetic , Wood , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Transformed , Chromatin Assembly and Disassembly , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , Humans , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Receptors, Aryl Hydrocarbon/genetics , Repressor Proteins/genetics
2.
Toxicology ; 461: 152897, 2021 09.
Article in English | MEDLINE | ID: mdl-34403729

ABSTRACT

Cytochrome P450 family 1 (CYP1) enzymes contribute both to metabolism of xenobiotics and to the control of endogenous levels of ligands of the aryl hydrocarbon receptor (AhR). Their activities, similar to other CYPs, can be altered in tumor tissues. Here, we examined a possible role of proliferative/survival pathways signaling, which is often deregulated in tumor cells, and possible links with p300 histone acetyltransferase (a transcriptional co-activator) in the control of CYP1 expression, focusing particularly on CYP1A1. Using cell models derived from human liver, we observed that the induction of CYP1A1 expression, as well as other CYP1 enzymes, was reduced in exponentially growing cells, as compared with their non-dividing counterparts. The siRNA-mediated inhibition of proliferation/pro-survival signaling pathway effectors (such as ß-catenin and/or Hippo pathway effectors YAP/TAZ) increased the AhR ligand-induced CYP1A1 mRNA levels in liver HepaRG cells, and/or in colon carcinoma HCT-116 cells. The activation of proliferative Wnt/ß-catenin signaling in HCT-116 cells reduced both the induction of CYP1 enzymes and the binding of p300 to the promoter of CYP1A1 or CYP1B1 genes. These results seem to indicate that aberrant proliferative signaling in tumor cells could suppress induction of CYP1A1 (or other CYP1 enzymes) via competition for p300 binding. This mechanism could be involved in modulation of the metabolism of both endogenous and exogenous substrates of CYP1A1 (and other CYP1 enzymes), with possible further consequences for alterations of the AhR signaling in tumor cells, or additional functional roles of CYP1 enzymes.


Subject(s)
Cell Proliferation/physiology , Colonic Neoplasms/pathology , Cytochrome P-450 CYP1A1/genetics , Liver/pathology , Cell Line, Tumor , Cell Survival/physiology , Colonic Neoplasms/genetics , Cytochrome P-450 CYP1A1/biosynthesis , E1A-Associated p300 Protein/metabolism , Enzyme Induction/physiology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Hippo Signaling Pathway/physiology , Humans , Signal Transduction/physiology , Wnt Signaling Pathway/physiology
3.
Toxicol Lett ; 349: 155-164, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34171359

ABSTRACT

Cytochrome P450 1A1 (CYP1A1) is a member of a subfamily of enzymes involved in the metabolism of both endogenous and exogenous substrates and the chemical activation of xenobiotics to carcinogenic derivatives. Here, the effects of nicotine, a major psychoactive compound present in cigarette smoke, on CYP1A1 expression and human hepatocellular carcinoma (HepG2) cell proliferation were investigated. Nicotine stimulated CYP1A1 expression via the transcription factors, activator protein 1, nuclear factor-kappa B, and the aryl hydrocarbon receptor (AhR) signaling pathway. Pharmacological inhibition and mutagenesis studies indicated that p38 mitogen-activated protein kinase, as well as RelA (or p65), mediated the upregulation of CYP1A1 of nicotine in HepG2 cells. The antioxidant compound, N-acetyl-cysteine, abrogated nicotine-activated production of reactive oxygen species and inhibited CYP1A1 expression by nicotine. Furthermore, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity was inhibited by diphenyleneiodonium (an NADPH oxidase inhibitor). Thus, these results demonstrated that AhR played an important role in nicotine-induced CYP1A1 expression. Additionally, liver hepatocellular carcinoma HepG2 cells treated with nicotine exhibited markedly enhanced proliferation via CYP1A1 expression and Akt activation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP1A1/biosynthesis , Liver Neoplasms/enzymology , Nicotine/toxicity , Nicotinic Agonists/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factor AP-1/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Appl Toxicol ; 41(9): 1380-1389, 2021 09.
Article in English | MEDLINE | ID: mdl-33569802

ABSTRACT

Glia cells provide supportive functions to the central nervous system and can be compromised by environmental contaminants. The primary objective of this study was to characterize the effects of in vitro exposure to perfluorooctanoic acid, a persistent environmental contaminant and/or monocrotophos (MCP), a neurotoxic organophosphate that is rapidly metabolized, to astroglia SVG p12 cells. The endpoints evaluated include cell viability, intracellular glutamate levels as a marker of astrocyte homeostasis function, differential gene expression for selected proteins, which include inflammatory markers (tachykinin), astrocytosis (nestin), S100B, and metabolism enzymes (CYP1A1). The results from cell viability revealed significant differences from the controls at some of the concentrations tested. Also, intracellular glutamate levels were elevated at the 10-µM concentration for perfluorooctanoic acid (PFOA) as well as the 10-µM PFOA/5-µM MCP concentration. Gene expression results at 80-µM PFOA concentration revealed a significant increase in the expression of S100B, tachykinin and CYP1A1. A combination of 10-µM PFOA/20-µM MCP caused a significant decrease in the expression of tachykinin. Gene expression for MCP exposures produced a decrease at the 20-µM MCP concentration. Immunofluorescence results indicated an increase in nestin protein expression for the 20-µM concentration of MCP, which contradicted the gene expression at the same concentration tested. The results indicate that toxicity to glia cells can compromise critical glia functions and could be implicated in neurodegenerative diseases.


Subject(s)
Astrocytes/drug effects , Caprylates/toxicity , Fluorocarbons/toxicity , Insecticides/toxicity , Monocrotophos/toxicity , Animals , Cell Line , Cell Survival/drug effects , Cytochrome P-450 CYP1A1/biosynthesis , Environmental Pollutants/toxicity , Female , Gene Expression/drug effects , Glutamic Acid/metabolism , Homeostasis/drug effects , Humans , Nestin/biosynthesis , PC12 Cells , Pregnancy , Rats , S100 Calcium Binding Protein beta Subunit/biosynthesis , Tachykinins/biosynthesis
5.
J Biochem Mol Toxicol ; 35(4): e22695, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33393179

ABSTRACT

Benzo(a)pyrene [B(a)P], which is a carcinogen, is a substance most typically known in cigarette smoke and considered as an important intermediary of lung cancer. The enzyme CYP1A1 is crucial for the metabolic conversion of B(a)P into the intermediates that induce carcinogenesis. Stimulation of the aryl hydrocarbon receptor, which is regulated by B(a)P, is thought to induce numerous signaling cascades. Interruption in the mitogen-activated protein kinase (MAPK) pathway causes changes in cellular processes and may alter the AhR pathway. The aim of this investigation is to examine the potential ability of a flavonoid pinocembrin (PCB) to alleviate B(a)P toxicity and analyze the underlying molecular mechanisms. We found that PCB inhibited DNA adduct formation by attenuating CYP1A1 expression through the suppression of the AhR/Src/ERK pathways. PCB mitigated the B(a)P-stimulated DNA damage, inhibited Src and ERK1/2 expression, decreased CYP1A1 expression, and reduced the B(a)P-induced stimulation of NF-κB and MAPK signaling in lung epithelial cells. Finally, the activity of CYP1A1 and Src in lung tissues from mice supplemented with PCB was noticeably decreased and lower than that in lung tissues from mice supplemented with B(a)P alone. Collectively, these data suggest that PCB may alleviate the toxic effects of PAHs, which are important environmental pollutants.


Subject(s)
Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/biosynthesis , Flavanones/pharmacology , Gene Expression Regulation, Developmental/drug effects , Lung/enzymology , MAP Kinase Signaling System/drug effects , Animals , Cell Line , Cytochrome P-450 CYP1A1/genetics , Female , Humans , Lung/pathology , Mice
6.
Sci Rep ; 10(1): 21891, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318545

ABSTRACT

The Cuvier's beaked whale (Ziphius cavirostris) is one of the least known cetacean species worldwide. The decreasing population trend and associated threats has led to the IUCN categorising the Mediterranean subpopulation as Vulnerable on the Red List of Threatened Species. This study aimed to investigate for the first time the ecotoxicological status of Cuvier's beaked whale in the NW Mediterranean Sea. The study sampled around the 20% of the individuals belonging to the Ligurian subpopulation, collecting skin biopsies from free-ranging specimens. The levels of polychlorinated biphenyl (PCBs), polybrominated diphenyl ethers (PBDEs) and induction of cytochrome's P450 (CYP1A1 and CYP2B isoforms) were evaluated. Results highlighted that the pattern of concentration for the target contaminants was PCBs > PBDEs and the accumulation values were linked to age and sex, with adult males showing significantly higher levels than juvenile. Concerns raised by the fact that 80% of the individuals had PCB levels above the toxicity threshold for negative physiological effects in marine mammals. Therefore, these findings shed light on this silent and serious threat never assessed in the Mediterranean Cuvier's beaked whale population, indicating that anthropogenic pressures, including chemical pollution, may represent menaces for the conservation of this species in the Mediterranean Sea.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Gene Expression Regulation, Enzymologic , Skin/enzymology , Whales/metabolism , Animals , Biopsy , Halogenated Diphenyl Ethers/toxicity , Isoenzymes/biosynthesis , Mediterranean Sea , Polychlorinated Biphenyls/toxicity , Skin/pathology , Water Pollution, Chemical
7.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374326

ABSTRACT

Previous preclinical studies have demonstrated the otoprotective effects of resveratrol (RV) at low doses. This study aimed to investigate the dose-dependent effects of RV in rats with cisplatin (CXP)-induced hearing loss. Sprague-Dawley rats (8-weeks old) were divided into six treatment groups (n = 12/group) and treated as follows: control, 0.5 mg/kg RV, 50 mg/kg RV, CXP, 0.5 mg/kg RV + CXP), and 50 mg/kg RV + CXP groups. CXP (3 mg/kg) was intraperitoneally injected for 5 days. RV (0.5 or 50 mg/kg) was intraperitoneally injected for 10 days from the first day of CXP administration. Auditory brainstem response (ABR) thresholds were measured before and within 3 days at the end of the drug administration. Cochlear tissues were harvested, and the outer hair cells were examined using cochlear whole mounts. The mRNA expression of NFκB, IL6, IL1ß, and CYP1A1, and protein levels of aryl hydrocarbon receptor (AhR) and cytosolic and nuclear receptor for advanced glycation endproducts (RAGE) were evaluated. The ABR threshold increased in the 50 mg/kg RV and CXP groups at 4, 8, 16, and 32 kHz. The 0.5 mg/kg RV + CXP group demonstrated decreased hearing thresholds at 4 and 32 kHz compared to the CXP group. Cochlear whole-mount analysis revealed loss of outer hair cells in the 50 mg/kg RV and CXP groups and partial prevention of these cells in the 0.5 mg/kg RV + CXP group. The mRNA expressions of NFκB, IL6, and IL1ß were increased in the 50 mg/kg RV and CXP groups compared to the control group. In contrast, these levels were decreased in the 0.5 mg/kg RV + CXP group compared to the CXP group. The mRNA expression of CYP1A1 was increased in the CXP group, while it was decreased in the 0.5 mg/kg RV + CXP group compared to the control group. The protein levels of AhR and cytosolic RAGE decreased in the 0.5 mg/kg RV group. Low-dose RV had partial otoprotective effects on CXP ototoxicity. The otoprotective effects of RV may be mediated through anti-oxidative (CYP1A1 and RAGE) and anti-inflammatory (NFκB, IL6, and IL1ß) responses. High-dose RV exerted an inflammatory response and did not ameliorate CXP-induced ototoxicity.


Subject(s)
Cisplatin/adverse effects , Hearing Loss, Sensorineural/drug therapy , Resveratrol/pharmacology , Animals , Antioxidants/pharmacology , Cochlea/drug effects , Cytochrome P-450 CYP1A1/biosynthesis , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Hearing Loss, Sensorineural/chemically induced , Inflammation , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , NF-kappa B p50 Subunit/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/metabolism
8.
Front Immunol ; 11: 2151, 2020.
Article in English | MEDLINE | ID: mdl-33013906

ABSTRACT

Objectives: Semaphorin 4D (Sema4D) is constitutively expressed on T cells and osteoclasts, and regulates T cell proliferation and bone remodeling. In addition, several studies have shown that Sema4D is involved in the pathogenesis of autoimmunity. We undertook this study to investigate the mechanism by which Sema4D affects the pathogenic progress of ankylosing spondylitis (AS). Methods: Soluble Sema4D (sSema4D) levels in serum were analyzed by enzyme-linked immunosorbent assay. The cell surface levels and transcripts of Sema4D were evaluated in CD4 + and CD19 + cells from the AS patients and healthy individuals. The mRNA expression levels were assessed by quantitative polymerase chain reaction (qPCR). The proportions of Treg cells and IL-17-producing T-cells (Th17 cells) differentiated from CD4 + T cells were analyzed by flow cytometric analysis. The aryl hydrocarbon receptor (AhR) agonistic effect of Sema4D was detected by analyzing the activation of downstream signaling pathways and target genes using Luciferase and EROD assay. Results: Levels of sSema4D were elevated in both serum from AS patients, and clinical features markers were correlated with serum sSema4D levels. Sema4D facilitated CD4 + T cells proliferation and Th17 cells differentiation and inhibited Treg cells differentiation by enhancing RORγt expression and reducing Foxp3 expression, with increasing expression and secretion of IL-17 and IL-22. It induced the expression and activity of AhR target gene CYP1A1 and XRE reporter activity via interaction with CD72. Conclusion: These findings indicate that Sema4D as a potent activator of T cells in the immune response contributes to the inflammation of AS by inducing imbalance in Th17 and Treg cell populations in an AhR-dependent manner, suggesting it is a crucial participant in AS pathogenesis.


Subject(s)
Antigens, CD/blood , CD4 Lymphocyte Count , Receptors, Aryl Hydrocarbon/agonists , Semaphorins/blood , Spondylitis, Ankylosing/immunology , T-Lymphocytes, Regulatory , Th17 Cells , Adult , Antigens, CD/pharmacology , Cell Differentiation , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Lymphocyte Activation , Male , Middle Aged , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Interleukin-17/biosynthesis , Receptors, Interleukin-17/genetics , Semaphorins/pharmacology , Spondylitis, Ankylosing/blood , Young Adult
9.
Toxicol Lett ; 331: 130-142, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32417428

ABSTRACT

Increasing production of corannulene (COR), a non-planar polycyclic aromatic hydrocarbon (PAH) with promising applications in many fields, has raised a concern about its potential toxic effects. However, no study has been undertaken to evaluate its metabolism and toxicity in mammals. In this study, the acute toxicities of COR in mice were compared with benzo[apyrene (BaP), a typical planar PAH with almost the same molecular weight. After 3-day exposures, the concentrations of COR in both plasma and tissues of mice were higher than that of BaP. However, blood chemistry and tissue weight monitoring showed no observable toxicities in COR-exposed mice. Compared to BaP, exposure to COR resulted in less activation of the aryl hydrocarbon receptor (AhR) and thus less induction of hepatic cytochrome P450 1A(CYP1A) enzymes, which play a critical role in metabolism of both COR and BaP. Additionally, COR also elicited less oxidative stress and microbiota alteration in the intestine than did BaP. RNA-seq analysis revealed that liver transcriptomes are responsive to COR and BaP, with less alterations observed in COR-exposed mice. Unlike BaP, exposure to COR had no effects on hepatic lipid and xenobiotic metabolism pathways. Nonetheless, COR appeared to alter the mRNA expressions of genes involved in carcinogenicity, oxidative stress, and immune-suppression. To conclude, this study for the first time unveils a comparative understanding of the acute toxic effects of COR to BaP in mice, and provides crucial insights into the future safety assessment of COR.


Subject(s)
Benzo(a)pyrene/toxicity , Intestines/drug effects , Liver/drug effects , Polycyclic Aromatic Hydrocarbons/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Administration, Oral , Animals , Benzo(a)pyrene/pharmacokinetics , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2/biosynthesis , Injections, Intraperitoneal , Intestines/pathology , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Mice, Inbred C57BL , Organ Size/drug effects , Polycyclic Aromatic Hydrocarbons/blood , Polycyclic Aromatic Hydrocarbons/pharmacokinetics , Rats, Sprague-Dawley , Superoxide Dismutase/blood , Tissue Distribution
10.
Aquat Toxicol ; 222: 105471, 2020 May.
Article in English | MEDLINE | ID: mdl-32199139

ABSTRACT

The toxicity of waterborne retene (7-isopropyl-1-methyl phenanthrene) to post-hatch embryos of rainbow trout (Oncorhynchus mykiss) was assessed at 5 and 11 °C. Survival times of retene-exposed embryos were 70 % longer at 5 °C than at 11 °C, but survival times and LC50 s did not vary when time was expressed as degree-days (thermal units), i.e., at a common stage of development. The size of survivors decreased with increasing retene concentrations, but not with temperature. Retene did not bioconcentrate to any extent (bioconcentration factors < 2) at either temperature, indicating effective biotransformation by embryos. However, concentrations of retene metabolites were slightly higher at 5 °C, suggesting slower excretion rates than at 11 °C. The relative expression of cytochrome P450 proteins (CYP1A) did not vary with temperature but increased with retene concentration, as indicated by cyp1a mRNA concentrations. The induction of CYP1A protein by retene exposure was evident in the vasculature of eye, brain, heart, kidney, liver, gill, mouth, intestine, muscle, and yolk-sac. However, immunohistochemical staining was greater at 5 than at 11 °C for all tissues except liver and muscle. Overall, temperature effects on retene toxicity disappeared when the duration of embryo development and retene exposure were expressed as thermal units (degree-days). Temperature controlled the rate of embryo development and the rate of toxicity (time to a toxic endpoint), but not the concentrations that were toxic.


Subject(s)
Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Oncorhynchus mykiss/metabolism , Phenanthrenes/toxicity , Temperature , Water Pollutants, Chemical/toxicity , Animals , Cytochrome P-450 CYP1A1/biosynthesis , Dose-Response Relationship, Drug , Embryo, Nonmammalian/metabolism , Enzyme Induction/drug effects , Lethal Dose 50
11.
Cell Immunol ; 349: 104047, 2020 03.
Article in English | MEDLINE | ID: mdl-32019673

ABSTRACT

The polarization of macrophages is critical to inflammation and tissue repair, with unbalanced macrophage polarization associated with critical dysfunctions of the immune system. Cytochrome P450 1A1 (CYP1A1) is a hydroxylase mainly controlled by the inflammation-limiting aryl hydrocarbon receptor (AhR), which plays a critical role in mycoplasma infection, oxidative stress injury, and cancer. Arginase-1 (Arg-1) is a surrogate for polarized alternative macrophages and is important to the production of nitric oxide (NO) by the modulation of arginine. In the present study, we found CYP1A1 to be upregulated in IL-4-stimulated mouse peritoneal macrophages (PMs) and human peripheral blood monocytes. Using CYP1A1-overexpressing RAW264.7 cells (CYP1A1/RAW) we found that CYP1A1 augmented Arg-1 expression by strengthening the activation of the JAK1/STAT6 signaling pathway in macrophages treated with IL-4. 15(S)-HETE, a metabolite of CYP1A1 hydroxylase, was elevated in IL-4-induced CYP1A1/RAW cells. Further, in macrophages, the loss-of-CYP1A1-hydroxylase activity was associated with reduced IL-4-induced Arg-1 expression due to impaired 15(S)-HETE generation. Of importance, CYP1A1 overexpressing macrophages reduced the inflammation associated with LPS-induced peritonitis. Taken together, these findings identified a novel signaling axis, CYP1A1-15(S)-HETE-JAK1-STAT6, that may be a promising target for the proper maintenance of macrophage polarization and may also be a means by which to treat immune-related disease due to macrophage dysfunction.


Subject(s)
Arginase/biosynthesis , Cytochrome P-450 CYP1A1/physiology , Janus Kinase 1/antagonists & inhibitors , Macrophages, Peritoneal/drug effects , Peritonitis/prevention & control , STAT6 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Adoptive Transfer , Animals , Arachidonate 15-Lipoxygenase/physiology , Arginase/genetics , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Endotoxins/toxicity , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/genetics , Hydroxyeicosatetraenoic Acids/pharmacology , Interleukin-4/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Peritonitis/chemically induced , RAW 264.7 Cells , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , THP-1 Cells , Up-Regulation/drug effects
12.
Toxicol Lett ; 321: 114-121, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31830554

ABSTRACT

Despite numerous studies on the toxicities of planar polycyclic aromatic hydrocarbons (PAHs), very little is known about the toxicological profiles of non-planar PAHs. In the present study, the cytotoxicity of corannulene (COR), a typical bowl-shaped PAH with a myriad of applications in the area of material chemistry, and benzo[a]pyrene (BaP), a typical planar PAH with similar molecular weight, were systematically compared in various cell lines. Compared with BaP, exposure to COR resulted in less cytotoxic responses in both human (HepG2) and murine (Hepa1-6) hepatoma cells, which was characterized with a slower cellular accumulation as well as a weaker induction of cytochrome P450 1 (CYP1/Cyp1) isozymes. Knockdown of aryl hydrocarbon receptor (AhR) by siRNA attenuated the inductive effect of COR on CYP1A/Cyp1a mRNA levels in these two cell lines. Further analysis revealed that derivatization greatly influenced the cytotoxicity of COR, which was positively correlated with their binding affinities to the AhR, as demonstrated by in silico molecular docking. Overall, these results suggest that AhR appears to be involved in the cytotoxic responses of COR and its derivatives, providing a fundamental understanding of the biological effects of bowl-like PAHs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/agonists , Hepatocytes/drug effects , Polycyclic Aromatic Hydrocarbons/toxicity , Receptors, Aryl Hydrocarbon/agonists , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Mice , Molecular Docking Simulation , Polycyclic Aromatic Hydrocarbons/metabolism , Protein Binding , Protein Conformation , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Structure-Activity Relationship
13.
Ecotoxicol Environ Saf ; 189: 110041, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31816496

ABSTRACT

Microplastic particles (MPs) from lipophilic polymers have been shown to efficiently accumulate hydrophobic organic contaminants (HOCs) in aquatic environments. MPs have, therefore, frequently been discussed as vectors for contaminants, enhancing HOC uptake by various organisms after ingestion followed by pollutant release; however, integrative models of sorption argue against this mechanism and even predict cleansing of pollutants from biological systems under particular circumstances. In order to experimentally investigate such a depuration mechanism, RTL-W1 cells were dosed with three 7-ethoxyresorufin-O-deethylase (EROD) inducers of distinct lipophilicity via the medium before adding both native and hexane-purified polyethylene MPs (20-25 µm) to the medium surface. EROD activity was significantly reduced in the presence of MP, the extent of which correlated with the inducers' lipophilicity (KOW) and thus affinity to MP. For hexane-purged MPs and TCDD (KOW = 6.8), MPs reduce the bioavailability by up to 79%; the effect was marginally weaker with benzo[k]fluoranthene (KOW = 6.11) and almost absent with ß-Naphthoflavone (KOW = 4.68). Compared to hexane-purged MPs, native particles possessed slightly less detoxification potential. These experimental results corroborate theoretically predicted mechanisms of detoxification via MPs. Yet, it is unclear if, under corresponding conditions in the environment, MPs can compete with organismal tissues for highly lipophilic compounds and, if so, to which degree they may act as a sink reducing the amount of bioavailable pollutants in situ. However, the present results suggest that in scenarios where pollutant-free MPs interact with organisms that accumulated HOCs via other routes of uptake, qualitatively the presence of such a mechanism is likely.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Enzyme Induction/drug effects , Microplastics/pharmacology , Water Pollutants, Chemical/pharmacology , Animals , Cell Line , Cytochrome P-450 Enzyme Inducers/pharmacology , Fluorenes/pharmacology , Hydrophobic and Hydrophilic Interactions , beta-Naphthoflavone/pharmacology
14.
Toxicol Lett ; 319: 85-94, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31730885

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a chronic hepatic disease associated with the excessive accumulation of lipids in the liver. Premenopausal women are protected from the liver metabolic complications of obesity compared with body mass index (BMI)-matched men. This protection may be related to estrogen's ability to limit liver fat accumulation. Aryl hydrocarbon receptor (AhR), a novel regulator of NAFLD, may be an important target for regulating estrogen homeostasis. In present study, we used benzo[a]pyrene (BaP), a classic and potent ligand of AhR, to activate AhR pathway causes overexpression of the estrogen-metabolizing enzyme cytochrome P450 1A1 (CYP1A1) and affects the expression of important genes involved in hepatic lipid regulation. BaP induces CYP1A1 expression through AhR signaling and inhibits the protective effect of 17ß-estradiol (E2) on hepatic steatosis, characterized by triglyceride accumulation, and markers of liver damage are significantly elevated. The expression of adipogenic genes involved in the hepatic lipid metabolism of sterol regulatory element-binding protein-1c (SREBP-1c) was increased compared with that in the control group. Furthermore, the mRNA and protein levels of peroxisome proliferator-activated receptor alpha (PPARα), which is involved in fatty acid oxidation, were significantly reduced. Taken together, our results revealed that the steatotic effect of AhR is likely due to overexpression of the E2 metabolic enzyme CYP1A1, which affects the estrogen signaling pathway, leading to the suppression of fatty acid oxidation, inhibition of the hepatic export of triglycerides, and an increase in peripheral fat mobilization. The results from this study may help establish AhR as a novel therapeutic and preventive target for fatty liver disease.


Subject(s)
Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Benzo(a)pyrene/pharmacology , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Estradiol/pharmacology , Estrogens/metabolism , Female , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , PPAR alpha/biosynthesis , PPAR alpha/genetics , Receptors, Aryl Hydrocarbon/agonists , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/biosynthesis , Sterol Regulatory Element Binding Protein 1/genetics , Triglycerides/metabolism
15.
Toxicol Lett ; 313: 66-76, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31201936

ABSTRACT

Mono-methylindoles (MMI) were described as agonists and/or antagonists of the human aryl hydrocarbon receptor (AhR). Here, we investigated the effects of MMI on AhR-CYP1A pathway in human hepatocytes and HepaRG cells derived from human progenitor hepatic cells. All MMI, except of 2-methylindole, strongly induced CYP1A1 and CYP1A2 mRNAs in HepaRG cells. Induction of CYP1A genes was absent in AhR-knock-out HepaRG cells. Consistently, CYP1A1 and CYP1A2 mRNAs and proteins were induced by all MMIs (except 2-methylindole), in human hepatocytes. The enzyme activity of CYP1A1 was inhibited by MMIs in human hepatocytes and LS180 colon cancer cells in a concentration-dependent manner (IC50 values from 1.2 µM to 23.8 µM and from 3.4 µM to 11.4 µM, respectively). Inhibition of CYP1A1 activity by MMI in human liver microsomes was much weaker as compared to that in intact cells. Incubation of parental MMI with human hepatocytes either diminished (4-methylindole, 6-methylindole) or enhanced (7-methylindole) their agonist effects on AhR in AZ-AHR reporter cells. In conclusion, overall effects of MMI on AhR-CYP1A pathway in human cells comprise the induction of CYP1A genes through AhR, the inhibition of CYP1A catalytic activity and possibly the metabolic transformation causing loss or gain of AhR agonist activity of parental compounds.


Subject(s)
Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Hepatocytes/drug effects , Indoles/pharmacology , Aged , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Colonic Neoplasms/enzymology , Cytochrome P-450 CYP1A1/genetics , Dose-Response Relationship, Drug , Enzyme Induction , Female , Hepatocytes/enzymology , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Middle Aged , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Young Adult
16.
Dokl Biochem Biophys ; 485(1): 150-152, 2019 Mar.
Article in English | MEDLINE | ID: mdl-31201638

ABSTRACT

A hybrid 6XRE-hTERT promoter consisting of the hTERT tumor-specific promoter and six copies of the XRE element from the CYP1A1 human gene promoter was created. Using a human lung cancer cells as a model, we showed that XRE elements in the hybrid promoter greatly increase the activity of the hTERT promoter and ensure the reporter gene transcriptional activation in response to the treatment of the cells with the AhR ligand benzo(a)pyrene. However, similar effects were also observed in normal human bronchial epithelial cells HBEpC, which indicates the loss of the tumor-specific activity by the 6XRE-hTERT hybrid promoter. XRE elements can be used for nonspecific transcription enhancement but are unsuitable for the creation of tumor-specific promoters with enhanced activity.


Subject(s)
Cytochrome P-450 CYP1A1 , Response Elements , Telomerase , Transcriptional Activation/drug effects , A549 Cells , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzo(a)pyrene/toxicity , Bronchi/metabolism , Bronchi/pathology , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Telomerase/biosynthesis , Telomerase/genetics
17.
Toxicol Sci ; 170(2): 310-319, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31086989

ABSTRACT

Environmental pollutants including halogenated aromatic hydrocarbons and polycyclic aromatic hydrocarbons, including benzo[a]pyrene, exert their deleterious effects through the activation of the aryl hydrocarbon receptor (AHR) and by the resulting transcription of genes not yet fully identified. Ligand-bound AHR translocates from cytoplasm to nucleus, where it dimerizes with the aryl hydrocarbon receptor nuclear translocator (ARNT) protein. The AHR/ARNT dimer binds to enhancer regions of responsive genes to activate transcription. AHR also mediates carcinogenesis caused by PAHs, likely via CYP1A1, CYP1A2, and CYP1B1, which are massively induced by activated AHR in many tissues and generate carcinogenic electrophilic derivatives of PAHs. In the current study, we have used the mouse GeCKOv2 genome-wide CRISPR/Cas9 library to identify novel genes in the AHR pathway by taking advantage of a B[a]P selection assay that we previously used to identify core AHR pathway genes in Hepa-1c1c7 murine hepatoma cells. Besides Ahr, Arnt, and Cyp1a1, we report the identification of multiple additional putative AHR pathway genes including several that we validated. These include cytochrome P450 reductase (Por), which mediates redox regeneration of cytochromes P450, and 5 genes of the heme biosynthesis pathway: delta-aminolevulinate synthase 1 (Alas1), porphobilinogen deaminase (Hmbs), uroporphyrinogen decarboxylase (Urod), coproporphyrinogen oxidase (Cpox), and ferrochelatase (Fech): heme being an essential prosthetic group of cytochrome P450 proteins. Notably, several of these genes were identified by GeCKO screening, despite not being identifiable by reverse genetics approaches. This indicates the power of high-sensitivity genome-wide genetic screening for identifying genes in the AHR pathway.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Cytochrome P-450 CYP1A1/biosynthesis , Receptors, Aryl Hydrocarbon/physiology , Animals , Benzo(a)pyrene/toxicity , Enzyme Induction , Heme/biosynthesis , Mice , Tumor Cells, Cultured
18.
Cardiovasc Toxicol ; 19(6): 565-574, 2019 12.
Article in English | MEDLINE | ID: mdl-31115867

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is associated with hypertension in humans and animals, and studies suggest that cytochrome P4501A1 (Cyp1a1) induction and vascular dysfunction may contribute. We investigated the role of perivascular adipose tissue (PVAT) and Cyp1a1 in TCDD-induced vascular dysfunction. Cyp1a1 wild-type (WT) and knockout (KO) male mice were fed a dough pill containing 1,4-p-dioxane (TCDD vehicle control) on days 0 and 7, or 1000 ng/kg TCDD on day 0 and 250 ng/kg TCDD on day 7. mRNA expression of Cyp1a1 was assessed on days 3, 7, and 14, and of Cyp1b1, 1a2, angiotensinogen, and phosphodiesterase 5a on day 14. Dose-dependent vasoconstriction to a thromboxane A2 mimetic (U46619), and vasorelaxation to acetylcholine and a nitric oxide donor (S-nitroso-N-acetyl-DL-penicillamine, SNAP), were investigated in the aorta with and without PVAT. Cyp1a1 and 1a2 mRNA was induced in aorta of WT mice only with PVAT, and Cyp1a1 induction was sustained through day 14. TCDD significantly enhanced constriction to U46619 in WT mice and inhibited relaxation to both acetylcholine and SNAP, but only in the presence of PVAT. The effects of TCDD on U46619 constriction and SNAP relaxation were not observed in Cyp1a1 KO mice. Finally, in aorta + PVAT of WT mice TCDD significantly induced expression of angiotensinogen and phosphodiesterase 5a both of which could contribute to the TCDD-induced vascular dysfunction. These data establish PVAT as a TCDD target which is critically involved in mediating vascular dysfunction. TCDD enhances vasoconstriction via the thromboxane/prostanoid (TP) receptor and inhibits vasorelaxation via nitric oxide (NO) signaling. This TCDD-induced vascular dysfunction requires perivascular adipose (PVAT) and cytochrome P4501a1 (CYP1a1) induction.


Subject(s)
Adipose Tissue/drug effects , Aorta/drug effects , Arterial Pressure/drug effects , Cytochrome P-450 CYP1A1/metabolism , Hypertension/chemically induced , Polychlorinated Dibenzodioxins/toxicity , Vasoconstriction/drug effects , Vasodilation/drug effects , Adipose Tissue/enzymology , Adipose Tissue/physiopathology , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Aorta/enzymology , Aorta/physiopathology , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/deficiency , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Time Factors
19.
Chem Res Toxicol ; 32(6): 1115-1122, 2019 06 17.
Article in English | MEDLINE | ID: mdl-30950278

ABSTRACT

Cytochrome P450s (CYPs), a superfamily of enzymes, are involved in the biotransformation of endogenous and xenobiotic chemicals and mainly responsible for the metabolic clearance of widely prescribed drugs. Out of the 57 human isoforms, only a few, most notably CYP3A4, are considered to be important in this process. CYP1A1, one of the three isoforms of the CYP1 family, is widely believed to play an important role in the metabolism and activation of numerous procarcinogens, e.g., polyaromatic hydrocarbons (PAHs) or aromatic amines. It is also known that CYP1A1 is highly inducible by endogenous and exogenous factors, e.g., PAHs. However, CYP1A1 has not been considered to play a significant role in the metabolic clearance of drugs, since this isoform has been detected only in extrahepatic tissues in small amounts. In contrast to conventional wisdom, we herein demonstrate the expression of CYP1A1 protein in human liver microsomal preparations. The expression levels of CYP1A1 were quantified by Western blot and LC/MS analyses and corresponded well with enzymatic activities of highly selective CYP1A1 reactions. In a panel of 29 individual liver microsomal preparations, highly variable and substantial expression levels (up to ∼10 pmol/mg) were measured. Together with the high selectivity and especially the high metabolic efficiency of CYP1A1 shown for granisetron and riociguat, it is demonstrated that CYP1A1 plays an important role in the metabolic clearance of these drugs and is responsible for the clinically observed interindividual variability in their pharmacokinetics. Therefore, the importance of CYP1A1 in drug discovery and development needs to be reconsidered.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Granisetron/pharmacokinetics , Liver/drug effects , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Blotting, Western , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A1/analysis , Humans , Liver/metabolism , Mass Spectrometry , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism
20.
Physiol Behav ; 204: 58-64, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30769105

ABSTRACT

Previous studies have shown that several aryl hydrocarbon receptor (AHR) agonists, including ß-naphthoflavone (BNF), elicit avoidance of novel food items in rodents, with this behavioral response displaying a similar dose-response to hepatic induction of CYP1A1. The avoidance has been found to bear substantial similarity to conditioned taste avoidance/aversion (CTA). The present study set out to confirm the indispensability of AHR in the avoidance response, to verify whether vagal afferent fibers are involved in it, and to see if AHR signaling might interfere with the effect of the classic trigger of CTA, LiCl. To this end, globally AHR deficient (AHRKO) or vagotomized wildtype rats were treated by gavage with 60 mg/kg BNF or ip with 0.15 M LiCl (4 ml/kg), and presented with chocolate which was either novel or familiar to them. Both the avoidance response and Cyp1a1 induction were missing in AHRKO rats. In contrast, Ahr+/- rats exhibited them in full, save for a single outlier. Total subdiaphragmatic vagotomy failed to interfere with the avoidance of novel or familiar chocolate or induction of Cyp1a1. After LiCl administration, male AHRKO rats showed a significantly mitigated suppression of chocolate consumption compared with wildtype animals (~60% vs. ~10% of control chocolate intake, respectively). A similar tendency was seen in females, but they were less responsive to LiCl. These findings corroborate AHR as a prerequisite of the BNF-induced novel food avoidance, prove vagal afferents unlikely mediators of this response, and imply an unforeseen involvement of AHR signaling in the thoroughly-characterized CTA instigated by LiCl.


Subject(s)
Avoidance Learning/drug effects , Basic Helix-Loop-Helix Transcription Factors/physiology , Feeding Behavior/drug effects , Lithium Chloride/pharmacology , Receptors, Aryl Hydrocarbon/physiology , Taste/drug effects , beta-Naphthoflavone/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction/drug effects , Gene Knockout Techniques , Neurons, Afferent/physiology , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/genetics , Vagotomy
SELECTION OF CITATIONS
SEARCH DETAIL
...