Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.426
Filter
1.
Cell ; 182(4): 886-900.e17, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32783918

ABSTRACT

Checkpoint immunotherapy unleashes T cell control of tumors, but is undermined by immunosuppressive myeloid cells. TREM2 is a myeloid receptor that transmits intracellular signals that sustain microglial responses during Alzheimer's disease. TREM2 is also expressed by tumor-infiltrating macrophages. Here, we found that Trem2-/- mice are more resistant to growth of various cancers than wild-type mice and are more responsive to anti-PD-1 immunotherapy. Furthermore, treatment with anti-TREM2 mAb curbed tumor growth and fostered regression when combined with anti-PD-1. scRNA-seq revealed that both TREM2 deletion and anti-TREM2 are associated with scant MRC1+ and CX3CR1+ macrophages in the tumor infiltrate, paralleled by expansion of myeloid subsets expressing immunostimulatory molecules that promote improved T cell responses. TREM2 was expressed in tumor macrophages in over 200 human cancer cases and inversely correlated with prolonged survival for two types of cancer. Thus, TREM2 might be targeted to modify tumor myeloid infiltrates and augment checkpoint immunotherapy.


Subject(s)
Immunotherapy , Membrane Glycoproteins/metabolism , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , CX3C Chemokine Receptor 1/metabolism , Cell Line, Tumor , Disease Models, Animal , Humans , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Methylcholanthrene/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/chemically induced , Neoplasms/pathology , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Tumor Microenvironment
2.
Cell Biol Toxicol ; 40(1): 5, 2024 01 25.
Article in English | MEDLINE | ID: mdl-38267663

ABSTRACT

3-Methylcholanthracene (3-MC) is one of the most carcinogenic polycyclic aromatic hydrocarbons (PAHs). Long-term exposure to PAHs has been thought of as an important factor in urothelial tumorigenesis. N6-methyladenosine (m6A) exists widely in eukaryotic organisms and regulates the expression level of specific genes by regulating mRNA stability, translation efficiency, and nuclear export efficiency. Currently, the potential molecular mechanisms that regulate m6A modification for 3-MC carcinogenesis remain unclear. Here, we profiled mRNA, m6A, translation and protein level using "-omics" methodologies, including transcriptomes, m6A profile, translatomes, and proteomics in 3-MC-transformed urothelial cells and control cells. The key molecules SLC3A2/SLC7A5 were screened and identified in 3-MC-induced uroepithelial transformation. Moreover, SLC7A5/SLC3A2 promoted uroepithelial cells malignant phenotype in vitro and in vivo. Mechanically, METTL3 and ALKBH5 mediated m6A modification of SLC3A2/SLC7A5 mRNA in 3-MC-induced uroepithelial transformation by upregulating the translation of SLC3A2/SLC7A5. Furthermore, programmable m6A modification of SLC3A2/SLC7A5 mRNA affected the expression of its proteins. Taken together, our results revealed that the m6A modification-mediated SLC3A2/SLC7A5 translation promoted 3-MC-induced uroepithelial transformation, suggesting that targeting m6A modification of SLC3A2/SLC7A5 may be a potential therapeutic strategy for bladder cancer related to PAHs.


Subject(s)
Large Neutral Amino Acid-Transporter 1 , Polycyclic Aromatic Hydrocarbons , Humans , Methylcholanthrene/toxicity , Carcinogenesis , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , RNA, Messenger/genetics , Methyltransferases/genetics , Fusion Regulatory Protein 1, Heavy Chain
3.
Int J Mol Sci ; 24(6)2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36982641

ABSTRACT

3-methylcholanthrene (3-MC) is a highly toxic environmental pollutant that impairs animal health. 3-MC exposure can cause abnormal spermatogenesis and ovarian dysfunction. However, the effects of 3-MC exposure on oocyte maturation and embryo development remain unclear. This study revealed the toxic effects of 3-MC exposure on oocyte maturation and embryo development. 3-MC with different concentrations of 0, 25, 50, and 100 µM was applied for in vitro maturation of porcine oocytes. The results showed that 100 µM 3-MC significantly inhibited cumulus expansion and the first polar body extrusion. The rates of cleavage and blastocyst of embryos derived from 3-MC-exposed oocytes were significantly lower than those in the control group. Additionally, the rates of spindle abnormalities and chromosomal misalignments were higher than those in the control group. Furthermore, 3-MC exposure not only decreased the levels of mitochondria, cortical granules (CGs), and acetylated α-Tubulin, but also increased the levels of reactive oxygen species (ROS), DNA damage, and apoptosis. The expression of cumulus expansion and apoptosis-related genes was abnormal in 3-MC-exposed oocytes. In conclusion, 3-MC exposure disrupted the nuclear and cytoplasmic maturation of porcine oocytes through oxidative stress.


Subject(s)
Methylcholanthrene , Oogenesis , Animals , Swine , Methylcholanthrene/pharmacology , Oocytes/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Embryonic Development , In Vitro Oocyte Maturation Techniques
4.
Eur J Immunol ; 51(6): 1473-1481, 2021 06.
Article in English | MEDLINE | ID: mdl-33684223

ABSTRACT

Therapeutic antibodies blocking PD-1-/PD-L1 interaction have achieved remarkable clinical success in cancer. In addition to blocking a target molecule, some isotypes of antibodies can activate complement, NK cells or phagocytes, resulting in death of the cell expressing the antibody's target. Human anti-PD-1 therapeutics use antibody isotypes designed to minimize such antibody-dependent lysis. In contrast, anti-PD-1 reagents used in mice are derived from multiple species, with different isotypes, and are not engineered to reduce target cell death: few studies analyze or discuss how antibody species and isotype may impact data interpretation. We demonstrate here that anti-PD-1 therapy to promote activation and proliferation of murine PD-1-expressing CD8 T cells sometimes led instead to a loss of antigen specific cells. This phenomenon was seen in two tumor models and a model of virus infection, and varied with the clone of anti-PD-1 antibody. Additionally, we compared competition among anti-PD-1 clones to find a combination that allows detection of PD-1-expressing cells despite the presence of blocking anti-PD1 antibodies in vivo. These data bring attention to the possibility of unintended target cell depletion with some commonly used anti-mouse PD-1 clones, and should provide a valuable resource for the design and interpretation of anti-PD-1 studies in mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Muromegalovirus/physiology , Sarcoma/immunology , Skin Neoplasms/immunology , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Death , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Drug Evaluation, Preclinical , Herpesviridae Infections/therapy , Humans , Immunoglobulin G/metabolism , Immunoglobulin Isotypes/metabolism , Methylcholanthrene , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Rats , Sarcoma/therapy , Skin Neoplasms/therapy
5.
Biol Pharm Bull ; 45(6): 793-797, 2022.
Article in English | MEDLINE | ID: mdl-35650105

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates the toxicity of dioxins and polycyclic aromatic hydrocarbons. Recent studies have suggested that AhR is involved in cancer immunity. In the present study, we examined whether AhR regulates the expression of immune checkpoint genes in breast cancer cells. We discovered that the mRNA expression of V-set domain containing T cell activation inhibitor 1 (VTCN1) that negatively regulates T cell immunity was upregulated by AhR agonists in breast cancer cell lines, MCF-7 and T47D. Furthermore, AhR knockout or knockdown experiments clearly demonstrated that upregulation of VTCN1 gene expression by 3-methylcholanthrene was AhR dependent. Luciferase reporter and chromatin immunoprecipitation assays revealed that this upregulation of VTCN1 gene expression was induced by the recruitment of AhR to the AhR responsive element in the VTCN1 gene promoter in MCF-7 cells. Taken together, AhR directly regulates VTCN1 gene expression in MCF-7 cells.


Subject(s)
Breast Neoplasms , Receptors, Aryl Hydrocarbon , V-Set Domain-Containing T-Cell Activation Inhibitor 1 , Basic Helix-Loop-Helix Transcription Factors , Breast Neoplasms/genetics , Female , Gene Expression , Humans , MCF-7 Cells , Methylcholanthrene/toxicity , Receptors, Aryl Hydrocarbon/genetics , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
6.
Biochem Biophys Res Commun ; 570: 131-136, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34280616

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates various toxicological and biological functions. We reported previously that 3-methylcholanthrene (3MC), an exogenous AhR agonist, inhibited tumorsphere (mammosphere) formation from breast cancer cell lines, while the endogenous AhR agonist, indirubin, very weakly inhibited this process. However, the difference in inhibition mechanism of mammosphere formation by 3MC or indirubin is still unknown. In this study, we established AhR-re-expressing (KOTR-AhR) cells from AhR knockout MCF-7 cells using the tetracycline (Tet)-inducible gene expression systems. To identify any difference in inhibition of mammosphere formation by 3MC or indirubin, RNA-sequencing (RNA-seq) experiments were performed using KOTR-AhR cells. RNA-seq experiments revealed that cell division cycle 20 (CDC20), which regulates the cell cycle and mitosis, was decreased by 3MC, but not by indirubin, in the presence of AhR expression. Furthermore, the mRNA and protein levels of CDC20 were decreased by 3MC in MCF-7 cells via the AhR. In addition, mammosphere formation was suppressed by small interfering RNA-mediated CDC20 knockdown compared to the negative control in MCF-7 cells. These results suggest that AhR activation by 3MC suppresses mammosphere formation via downregulation of CDC20 expression in breast cancer cells. This study provides useful information for the development of AhR-targeted anti-cancer drugs.


Subject(s)
Breast Neoplasms/genetics , Cdc20 Proteins/metabolism , Down-Regulation , Methylcholanthrene/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Spheroids, Cellular/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Transcriptome/genetics
7.
BMC Cancer ; 21(1): 629, 2021 May 28.
Article in English | MEDLINE | ID: mdl-34044797

ABSTRACT

BACKGROUND: Despite considerable medical proceedings, cancer is still a leading cause of death. Major problems for tumor therapy are chemoresistance as well as toxic side effects. In recent years, the additional treatment with the antidiabetic drug metformin during chemotherapy showed promising results in some cases. The aim of this study was to develop an in vitro tumor therapy model in order to further investigate the potential of a combined chemotherapy with metformin. METHODS: Cytotoxic effects of a combined treatment on BALB/c fibroblasts were proven by the resazurin assay. Based on the BALB/c cell transformation assay, the BALB/c tumor therapy model was established successfully with four different and widely used chemotherapeutics from different categories. Namely, Doxorubicin as a type-II isomerase inhibitor, Docetaxel as a spindle toxin, Mitomycin C as an alkylating agent and 5-Fluorouracil as an antimetabolite. Moreover, glucose consumption in the medium supernatant was measured and protein expressions were determined by Western Blotting. RESULTS: Initial tests for the combined treatment with metformin indicated unexpected results as metformin could partly mitigate the cytotoxic effects of the chemotherapeutic agents. These results were further confirmed as metformin induced resistance to some of the drugs when applied simultaneously in the tumor therapy model. Mechanistically, an increased glucose consumption was observed in non-transformed cells as well as in the mixed population of malignant transformed cell foci and non-transformed monolayer cells, suggesting that metformin could also increase glucose consumption in transformed cells. CONCLUSION: In conclusion, this study suggests a cautious use of metformin during chemotherapy. Moreover, the BALB/c tumor therapy model offers a potent tool for further mechanistic studies of drug-drug interactions during cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Metformin/pharmacology , Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BALB 3T3 Cells , Carcinogens/toxicity , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Culture Media/metabolism , Docetaxel/pharmacology , Docetaxel/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Evaluation, Preclinical , Drug Interactions , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Glucose/metabolism , Humans , Metformin/therapeutic use , Methylcholanthrene/toxicity , Mice , Mitomycin/pharmacology , Mitomycin/therapeutic use
8.
Int J Mol Sci ; 22(24)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34948000

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) accounts for 25% of cutaneous malignancies diagnosed in Caucasian populations. Surgical removal in combination with radiation and chemotherapy are effective treatments for cSCC. Nevertheless, the aggressive metastatic forms of cSCC still have a relatively poor patient outcome. Studies have linked actin cytoskeletal dynamics and the Wnt/ß-catenin signaling pathway as important modulators of cSCC pathogenesis. Previous studies have also shown that the actin-remodeling protein Flightless (Flii) is a negative regulator of cSCC. The aim of this study was to investigate if the functional effects of Flii on cSCC involve the Wnt/ß-catenin signaling pathway. Flii knockdown was performed using siRNA in a human late stage aggressive metastatic cSCC cell line (MET-1) alongside analysis of Flii genetic murine models of 3-methylcholanthrene induced cSCC. Flii was increased in a MET-1 cSCC cell line and reducing Flii expression led to fewer PCNA positive cells and a concomitant reduction in cellular proliferation and symmetrical division. Knockdown of Flii led to decreased ß-catenin and a decrease in the expression of the downstream effector of ß-catenin signaling protein SOX9. 3-Methylcholanthrene (MCA)-induced cSCC in Flii overexpressing mice showed increased markers of cancer metastasis including talin and keratin-14 and a significant increase in SOX9 alongside a reduction in Flii associated protein (Flap-1). Taken together, this study demonstrates a role for Flii in regulating proteins involved in cSCC proliferation and tumor progression and suggests a potential role for Flii in aggressive metastatic cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Microfilament Proteins/genetics , Skin Neoplasms/genetics , Trans-Activators/genetics , Up-Regulation , Wnt Signaling Pathway , Animals , Carcinoma, Squamous Cell/chemically induced , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Methylcholanthrene/adverse effects , Mice , Skin Neoplasms/chemically induced
9.
Bratisl Lek Listy ; 121(5): 325-330, 2020.
Article in English | MEDLINE | ID: mdl-32356428

ABSTRACT

BACKGROUND: Skin is the body's first defence against direct exposure to variety of chemicals. Polycyclic aromatic hydrocarbons such as 3-methylcholanthrene (3-MC) are common in polluted urban air and have a potential of producing harmful effects. Moreover, their late effects can occur months or years after exposure. OBJECTIVES: We aimed to investigate the long-term effects of 3-MC induced dermal toxicity on the expression of markers of apoptosis, pleiotropic cytokines, and oxidative stress and to determine the protective effect of cisplatin. METHODS: Groups were designed as control (group 1), 3-MC applied (group 2) and 3-MC+cisplatin applied mice (group 3). Cutaneous expressions of TGFß, PDGFA, PDGFC, bFGF, PDGFRα, USP28, and Ki67 were evaluated with qPCR. Total oxidant (TOS), total antioxidant (TAS) and oxidative stress index (OSI) values were determined in liver and kidney tissues. RESULTS: The expression levels of TGFß, PDGFRα, USP-28, Ki67, and PDGFA were decreased significantly in MC applied groups. Renal TAS levels were significantly lower in group-3. Liver and kidney OSI values were increased in both groups 2 and 3. CONCLUSION: The results indicated that low dose 3-MC caused oxidative stress and downregulated apoptotic and cytokine markers in the long term and cisplatin had no ameliorative effects on this degeneration processes (Tab. 3, Fig. 3, Ref. 32). Text in PDF www.elis.sk.


Subject(s)
DNA Damage , Methylcholanthrene , Oxidative Stress , Animals , Antioxidants , Biomarkers , Methylcholanthrene/toxicity , Mice
10.
Drug Metab Dispos ; 47(3): 279-282, 2019 03.
Article in English | MEDLINE | ID: mdl-30573465

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are environmental pollutants that activate the aryl hydrocarbon receptor, thereby triggering a range of biologic responses, exemplified by the induction of CYP1A1 PAHs can also regulate the expression of members of the CYP3A subfamily, with reports of mainly suppressive effects on mouse hepatic Cyp3a11 expression, but paradoxically both inductive and suppressive effects on human hepatic CYP3A4 expression. Understanding the regulation of CYP3A4 expression by PAHs is important because of the widespread exposure of humans to these chemicals and the central role of the CYP3A4 enzyme in the metabolism of clinically important drugs and endogenous substances. The present study used 3-methylcholanthrene (MC) as a model PAH to characterize the in vivo regulation of CYP3A4 expression and activity in humanized pregnane X receptor-constitutive androstane receptor-CYP3A4/3A7 mice. Adult mice were treated by intraperitoneal injection with MC (80 mg/kg), or corn oil vehicle, and euthanized 24 or 72 hours later. As a positive control response, pronounced induction of hepatic Cyp1a1 by MC was confirmed at both time points in males and females at the mRNA, protein, and catalytic activity levels. Basal hepatic CYP3A4 expression and activity were significantly higher in female versus male mice. MC treatment suppressed hepatic CYP3A4 in female mice at 72 hours postdosing at the mRNA, protein, and catalytic activity levels. A similar response was observed in male mice, although the suppression of CYP3A4 protein levels did not achieve statistical significance. This mouse model will facilitate further studies of the mechanisms and consequences of CYP3A4 suppression by PAHs.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Environmental Pollutants/toxicity , Gene Expression Regulation/drug effects , Methylcholanthrene/toxicity , Animals , Constitutive Androstane Receptor , Cytochrome P-450 CYP3A/genetics , Environmental Pollutants/administration & dosage , Female , Injections, Intraperitoneal , Liver/drug effects , Liver/metabolism , Male , Methylcholanthrene/administration & dosage , Mice , Mice, Transgenic , Models, Animal , Pregnane X Receptor/genetics , Pregnane X Receptor/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sex Factors
11.
Lipids Health Dis ; 18(1): 203, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31757214

ABSTRACT

BACKGROUND: A healthy gastric mucosal epithelium exhibits tumor-suppressive properties. Gastric epithelial cell dysfunction contributes to gastric cancer development. Oxysterols provided from food or cholesterol oxidation in the gastric epithelium may be further sulfated by hydroxysteroid sulfotransferase 2B1 (SULT2B1), which is highly abundant in the gastric epithelium. However, the effects of SULT2B1 on gastric epithelial function and gastric carcinogenesis are unclear. METHODS: A mouse gastric tumor model was established using carcinogenic agent 3-methylcholanthrene (3-MCA). A SULT2B1 deletion (SULT2B1-/-) human gastric epithelial line GES-1 was constructed by CRISPR/CAS9 genome editing system. RESULTS: The gastric tumor incidence was higher in the SULT2B1-/- mice than in the wild-type (WT) mice. In gastric epithelial cells, adenovirus-mediated SULT2B1b overexpression reduced the levels of oxysterols, such as 24(R/S),25-epoxycholesterol (24(R/S),25-EC) and 27-hydroxycholesterol (27HC). This condition also increased PI3K/AKT signaling to promote gastric epithelial cell proliferation, epithelization, and epithelial development. However, SULT2B1 deletion or SULT2B1 knockdown suppressed PI3K/AKT signaling, epithelial cell epithelization, and wound healing and induced gastric epithelial cell malignant transition upon 3-MCA induction. CONCLUSIONS: The abundant SULT2B1 expression in normal gastric epithelium might maintain epithelial function via the PI3K/AKT signaling pathway and suppress gastric carcinogenesis induced by a carcinogenic agent.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Stomach Neoplasms/genetics , Sulfotransferases/genetics , Animals , Base Sequence , CRISPR-Cas Systems , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/pathology , Gene Editing , Humans , Hydroxycholesterols/metabolism , Methylcholanthrene/administration & dosage , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Stomach Neoplasms/chemically induced , Stomach Neoplasms/enzymology , Stomach Neoplasms/mortality , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/deficiency , Survival Analysis
12.
Int J Mol Sci ; 20(9)2019 May 10.
Article in English | MEDLINE | ID: mdl-31083300

ABSTRACT

TCDD-inducible poly-ADP-ribose polymerase (TIPARP) is an aryl hydrocarbon receptor (AHR) target gene that functions as part of a negative feedback loop to repress AHR activity. Tiparp-/- mice exhibit increased sensitivity to the toxicological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), including lethal wasting syndrome. However, it is not known whether Tiparp-/- mice also exhibit increased sensitivity to other AHR ligands. In this study, we treated male Tiparp-/- or wild type (WT) mice with a single injection of 100 mg/kg 3-methylcholanthrene (3MC). Consistent with TIPARP's role as a repressor of AHR signaling, 3MC-treated Tiparp-/- mice exhibited increased hepatic Cyp1a1 and Cyp1b1 levels compared with WT mice. No 3MC-treated Tiparp-/- mice survived beyond day 16 and the mice exhibited chylous ascites characterized by an accumulation of fluid in the peritoneal cavity. All WT mice survived the 30-day treatment and showed no signs of fluid accumulation. Treated Tiparp-/- mice also exhibited a transient and mild hepatotoxicity with inflammation. 3MC-treated WT, but not Tiparp-/- mice, developed mild hepatic steatosis. Lipid deposits accumulated on the surface of the liver and other abdominal organs in the 3MC-Tiparp-/- mice. Our study reveals that Tiparp-/- mice have increased sensitivity to 3MC-induced liver toxicity, but unlike with TCDD, lethality is due to chylous ascites rather than wasting syndrome.


Subject(s)
Chylous Ascites/chemically induced , Chylous Ascites/enzymology , Methylcholanthrene/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Polychlorinated Dibenzodioxins/toxicity , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Azo Compounds/pharmacology , Chylous Ascites/pathology , Cytokines/metabolism , Fatty Liver/enzymology , Fatty Liver/pathology , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/pathology , Inflammation Mediators/metabolism , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Survival Analysis
13.
Carcinogenesis ; 39(7): 955-967, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29554273

ABSTRACT

The development of alternative methods to animal testing is a priority in the context of regulatory toxicology. Carcinogenesis is a field where the demand for alternative methods is particularly high. The standard rodent carcinogenicity bioassay requires a large use of animals, high costs, prolonged duration and shows several limitations, which can affect the comprehension of the human relevance of animal carcinogenesis. The cell transformation assay (CTA) has long been debated as a possible in vitro test to study carcinogenesis. This assay provides an easily detectable endpoint of oncotransformation, which can be used to anchor the exposure to the acquisition of the malignant phenotype. However, the current protocols do not provide information on either molecular key events supporting the carcinogenesis process, nor the mechanism of action of the test chemicals. In order to improve the use of this assay in the integrated testing strategy for carcinogenesis, we developed the transformics method, which combines the CTA and transcriptomics, to highlight the molecular steps leading to in vitro malignant transformation. We studied 3-methylcholanthrene (3-MCA), a genotoxic chemical able to induce in vitro cell transformation, at both transforming and subtransforming concentrations in BALB/c 3T3 cells and evaluated the gene modulation at critical steps of the experimental protocol. The results gave evidence for the potential key role of the immune system and the possible involvement of the aryl hydrocarbon receptor (AhR) pathway as the initial steps of the in vitro transformation process induced by 3-MCA, suggesting that the initiating events are related to non-genotoxic mechanisms.


Subject(s)
Carcinogens/toxicity , Cell Transformation, Neoplastic/chemically induced , Methylcholanthrene/toxicity , 3T3 Cells , Animals , Biological Assay , Carcinogenesis/chemically induced , Carcinogenicity Tests/methods , Mice , Mice, Inbred BALB C , Receptors, Aryl Hydrocarbon/metabolism
14.
Drug Metab Dispos ; 46(9): 1372-1381, 2018 09.
Article in English | MEDLINE | ID: mdl-29980579

ABSTRACT

CYP1A1 expression can be upregulated by the ligand-activated aryl hydrocarbon receptor (AHR). Based on prior observations with estrogen receptors and estrogen response elements, we tested the hypothesis that single-nucleotide polymorphisms (SNPs) mapping hundreds of base pairs (bp) from xenobiotic response elements (XREs) might influence AHR binding and subsequent gene expression. Specifically, we analyzed DNA sequences 5 kb upstream and downstream of the CYP1A1 gene for putative XREs. SNPs located ±500 bp of these putative XREs were studied using a genomic data-rich human lymphoblastoid cell line (LCL) model system. CYP1A1 mRNA levels were determined after treatment with varying concentrations of 3-methylcholanthrene (3MC). The rs2470893 (-1694G>A) SNP, located 196 bp from an XRE in the CYP1A1 promoter, was associated with 2-fold variation in AHR-XRE binding in a SNP-dependent fashion. LCLs with the AA genotype displayed significantly higher AHR-XRE binding and CYP1A1 mRNA expression after 3MC treatment than did those with the GG genotype. Electrophoretic mobility shift assay (EMSA) showed that oligonucleotides with the AA genotype displayed higher LCL nuclear extract binding after 3MC treatment than did those with the GG genotype, and mass spectrometric analysis of EMSA protein-DNA complex bands identified three candidate proteins, two of which were co-immunoprecipitated with AHR. In conclusion, we have demonstrated that the rs2470893 SNP, which maps 196 bp from a CYP1A1 promoter XRE, is associated with variations in 3MC-dependent AHR binding and CYP1A1 expression. Similar "distant SNP effects" on AHR binding to an XRE motif and subsequent gene expression might occur for additional AHR-regulated genes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cytochrome P-450 CYP1A1/genetics , Polymorphism, Single Nucleotide , Receptors, Aryl Hydrocarbon/metabolism , Xenobiotics/metabolism , 5' Untranslated Regions , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Cell Line, Tumor , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 Enzyme Inducers/pharmacology , Enzyme Induction , Humans , Methylcholanthrene/pharmacology , Protein Binding , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/genetics , Response Elements , Transcription, Genetic
15.
Biochim Biophys Acta Gen Subj ; 1862(3): 414-426, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29154903

ABSTRACT

BACKGROUND: Mesenchymal-to-epithelial transition (MET) is associated with altered cell adhesion patterns. Independent studies showed that cellular adhesion regulates low-dose hyper-radiosensitivity (HRS), a phenomenon reported widely in tumour cells. Therefore, present study aimed to investigate whether MET and associated cellular adhesion alterations affect cellular radiosensitivity. METHODS: We established multiple stages of MET by in vitro transformation of NIH3T3 mouse embryonic fibroblasts. Nutritional deprivation followed by repetitive treatment cycles of 3-methylcholanthrene and phorbol-12-myristate-13-acetate with frequent isolation of foci established three progressive strains (NIH3T3.1, NIH3T3x3, NIH3T3x8x3) depicting MET, and one strain (NIH3T3x12) with partial reversion. Alterations in morphology, cell adhesion properties, expression/intracellular localization of cell adhesion proteins, microRNA expression and cellular radiosensitivity were studied in these stably transformed cell strains. RESULTS: All four transformants had increased proliferation rate, saturation density, bipolarity, E-cadherin expression; coupled with reduced cell size/spreading, pseudopodia/migration, and fibroblast marker protein and vimentin. The most aggressive trans-differentiated (phenotypically epithelial) cell strain, NIH3T3x8x3 acquired ~30% higher growth potential associated with more than two-fold reduction in cell size and migration. These phenotypic changes accompanied ~40% reduction in endogenous or radiation-induced connexin-43 expression/mitochondrial translocation. Incidentally, all three progressive strains displayed prominent HRS (αs/αr: 7.95-37.29) whereas parental (NIH3T3) and reverting (NIH3T3x12) strains lacked HRS and had distinct radiation-induced Cx43 translocation into mitochondria. CONCLUSION: Our study shows that trans-differentiating fibroblasts progressively acquiring epithelial features during MET process, display low-dose hyper-radiosensitivity associated with altered Cx43 behaviour. GENERAL SIGNIFICANCE: This study demonstrates that MET progression triggers low-dose hyper-radiosensitivity in trans-differentiating cells, which has significant therapeutic implications.


Subject(s)
Cell Transdifferentiation/physiology , Connexin 43/metabolism , Fibroblasts/drug effects , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/metabolism , Radiation Tolerance/physiology , Animals , Cadherins/biosynthesis , Cell Adhesion , Cell Line, Transformed , Cell Movement , Cell Size , Cell Transformation, Neoplastic , Dose-Response Relationship, Radiation , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gamma Rays , Methylcholanthrene/toxicity , Mice , Mitochondria/metabolism , NIH 3T3 Cells , Phorbol Esters/toxicity , Protein Transport
16.
Nature ; 482(7385): 405-9, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22318517

ABSTRACT

Cancer immunoediting is a process by which immune cells, particularly lymphocytes of the adaptive immune system, protect the host from the development of cancer and alter tumour progression by driving the outgrowth of tumour cells with decreased sensitivity to immune attack. Carcinogen-induced mouse models of cancer have shown that primary tumour susceptibility is thereby enhanced in immune-compromised mice, whereas the capacity for such tumours to grow after transplantation into wild-type mice is reduced. However, many questions about the process of cancer immunoediting remain unanswered, in part because of the known antigenic complexity and heterogeneity of carcinogen-induced tumours. Here we adapted a genetically engineered, autochthonous mouse model of sarcomagenesis to investigate the process of cancer immunoediting. This system allows us to monitor the onset and growth of immunogenic and non-immunogenic tumours induced in situ that harbour identical genetic and histopathological characteristics. By comparing the development of such tumours in immune-competent mice with their development in mice with broad immunodeficiency or specific antigenic tolerance, we show that recognition of tumour-specific antigens by lymphocytes is critical for immunoediting against sarcomas. Furthermore, primary sarcomas were edited to become less immunogenic through the selective outgrowth of cells that were able to escape T lymphocyte attack. Loss of tumour antigen expression or presentation on major histocompatibility complex I was necessary and sufficient for this immunoediting process to occur. These results highlight the importance of tumour-specific-antigen expression in immune surveillance, and potentially, immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Gene Expression Regulation, Neoplastic , Immunologic Surveillance/immunology , Neoplasms/immunology , Animals , Antigens, Neoplasm/genetics , Disease Models, Animal , HEK293 Cells , Humans , Methylcholanthrene , Mice , Neoplasms/chemically induced , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Sarcoma/chemically induced , Sarcoma/genetics , Sarcoma/immunology , Sarcoma/pathology , T-Lymphocytes/immunology
17.
Nature ; 482(7385): 400-4, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22318521

ABSTRACT

Cancer immunoediting, the process by which the immune system controls tumour outgrowth and shapes tumour immunogenicity, is comprised of three phases: elimination, equilibrium and escape. Although many immune components that participate in this process are known, its underlying mechanisms remain poorly defined. A central tenet of cancer immunoediting is that T-cell recognition of tumour antigens drives the immunological destruction or sculpting of a developing cancer. However, our current understanding of tumour antigens comes largely from analyses of cancers that develop in immunocompetent hosts and thus may have already been edited. Little is known about the antigens expressed in nascent tumour cells, whether they are sufficient to induce protective antitumour immune responses or whether their expression is modulated by the immune system. Here, using massively parallel sequencing, we characterize expressed mutations in highly immunogenic methylcholanthrene-induced sarcomas derived from immunodeficient Rag2(-/-) mice that phenotypically resemble nascent primary tumour cells. Using class I prediction algorithms, we identify mutant spectrin-ß2 as a potential rejection antigen of the d42m1 sarcoma and validate this prediction by conventional antigen expression cloning and detection. We also demonstrate that cancer immunoediting of d42m1 occurs via a T-cell-dependent immunoselection process that promotes outgrowth of pre-existing tumour cell clones lacking highly antigenic mutant spectrin-ß2 and other potential strong antigens. These results demonstrate that the strong immunogenicity of an unedited tumour can be ascribed to expression of highly antigenic mutant proteins and show that outgrowth of tumour cells that lack these strong antigens via a T-cell-dependent immunoselection process represents one mechanism of cancer immunoediting.


Subject(s)
Exome/genetics , Exome/immunology , Immunologic Surveillance/immunology , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes/immunology , Algorithms , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Histocompatibility Antigens Class I/immunology , Humans , Male , Methylcholanthrene , Mice , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Models, Immunological , Neoplasms/chemically induced , Neoplasms/pathology , Reproducibility of Results , Sarcoma/chemically induced , Sarcoma/genetics , Sarcoma/immunology , Sarcoma/pathology
18.
Arch Toxicol ; 92(2): 907-919, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29094188

ABSTRACT

Daily exposure to low doses of 3-methylcholanthrene (3MC) during the pubertal period in rats disrupts both follicular growth and ovulation. Thus, to provide new insights into the toxicity mechanism of 3MC in the ovary, here we investigated the effect of daily exposure to 3MC on selected ovarian genes, the role of the aryl hydrocarbon receptor (AhR) and the level of epigenetic remodeling of histone post-transcriptional modifications. Immature rats were daily injected with 3MC (0.1 or 1 mg/kg) and mRNA expression of genes involved in different ovarian processes were evaluated. Of the 29 genes studied, 18 were up-regulated, five were down-regulated and six were not altered. To assess whether AhR was involved in these changes, we used the chromatin immunoprecipitation assay. 3MC increased AhR binding to promoter regions of genes involved in Notch signaling (Hes1, Jag1), activation of primordial follicles (Cdk2), cell adhesion (Icam1), stress and tumor progression (Dnajb6), apoptosis (Bax, Caspase-9) and expression of growth and transcription factors (Igf2, Sp1). Studying the trimethylation and acetylation of histone 3 (H3K4me3 and H3K9Ac, respectively) of these genes, we found that 3MC increased H3K4me3 in Cyp1a1, Jag1, Dnajb6, Igf2, Notch2, Adamts1, Bax and Caspase-9, and H3K9Ac in Cyp1a1, Jag1, Cdk2, Dnajb6, Igf2, Icam1, and Sp1. Co-treatment with α-naphthoflavone (αNF), a specific antagonist of AhR, prevented almost every 3MC-induced changes. Despite the low dose used in these experiments, daily exposure to 3MC induced changes in both gene expression and epigenomic remodeling, which may lead to premature ovarian failure.


Subject(s)
Benzoflavones/pharmacology , Methylcholanthrene/toxicity , Ovarian Follicle/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Acetylation , Animals , Chromatin Immunoprecipitation , Down-Regulation , Epigenesis, Genetic , Female , Histones/chemistry , Methylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Up-Regulation
19.
Ecotoxicol Environ Saf ; 161: 721-728, 2018 10.
Article in English | MEDLINE | ID: mdl-29940513

ABSTRACT

Many chemicals produced by human activities end up in the aquatic ecosystem causing adverse developmental and reproductive effects in aquatic organisms. There is evidence that some anthropogenic chemicals disturb bone formation and skeletal development but the lack of suitable in vitro and in vivo systems for testing has hindered the identification of underlying mechanisms of osteotoxicity. Several fish systems - an in vitro cell system to study extracellular matrix mineralization and in vivo systems to evaluate bone formation and skeletogenesis - were combined to collect data on the osteotoxic activity of 3-methylcholanthrene (3-MC), a polycyclic aromatic hydrocarbon. Anti-mineralogenic effects, increased incidence of skeletal deformities and reduced bone formation and regeneration were observed in zebrafish upon exposure to 3-MC. Pathway reporter array revealed the role of the aryl hydrocarbon receptor 2 (Ahr2) in the mechanisms underlying 3-MC osteotoxicity in mineralogenic cell lines. Analysis of gene expression in zebrafish larvae confirmed the role of Ahr2 in the signaling of 3-MC toxicity. It also indicated a possible complementary action of the pregnane X receptor (Pxr) in the regulation of genes involved in bone cell activity and differentiation but also in xenobiotic metabolism. Data reported here demonstrated the osteotoxicity of 3-MC but also confirmed the suitability of fish systems to gain insights into the toxic mechanisms of compounds affecting skeletal and bone formation.


Subject(s)
Methylcholanthrene/toxicity , Osteogenesis/drug effects , Animals , Calcification, Physiologic/drug effects , Cell Line , Humans , Larva/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Zebrafish/growth & development , Zebrafish/metabolism
20.
Andrologia ; 50(10): e13117, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30084500

ABSTRACT

Oestrus urine was proved as a potential endocrine modulator in alleviating the toxicity induced by 3-methylcholanthrene (3-MC) in male rats. We, in this study, aimed to prove the attributing potential of toxicity alleviation to squalene, an oestrus-specific pheromone in rats. A single dose of 3-methylcholanthrene (25 mg/kg BW, i.p.) was administered to male Wistar rats with concurrent exposure to squalene sprayed in bedding material (Group III). Group II rats did receive 3-MC treatment but did not expose to squalene. Group I rats were intact control neither administered 3-MC nor sprayed with squalene. In consequence of 3-MC toxicity, liver and testes weights were increased and the components of blood cells (RBC and WBC count, Hb level) and testosterone concentration were significantly reduced in Group II rats. Moreover, sperm count was reduced and antioxidants (testes and epididymis) were significantly altered. Exposure to squalene in Group III rats comparatively normalised all the variable components towards baseline and reorganised the histological architecture of reproductive tissues that were exacerbated with 3-MC toxicity. This study ultimately proved squalene as a potent molecule in alleviating the toxicity induced by 3-methylcholanthrene.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Sex Attractants/pharmacology , Squalene/pharmacology , Testis/drug effects , Animals , Blood Cell Count , Epididymis/drug effects , Liver/drug effects , Male , Methylcholanthrene/toxicity , Models, Animal , Rats , Rats, Wistar , Sperm Count , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL