Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(20)2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37895177

ABSTRACT

Melatonin is widely present in Nature. It has pleiotropic activities, in part mediated by interactions with high-affinity G-protein-coupled melatonin type 1 and 2 (MT1 and MT2) receptors or under extreme conditions, e.g., ischemia/reperfusion. In pharmacological concentrations, it is given to counteract the massive damage caused by MT1- and MT2-independent mechanisms. The aryl hydrocarbon receptor (AhR) is a perfect candidate for mediating the latter effects because melatonin has structural similarity to its natural ligands, including tryptophan metabolites and indolic compounds. Using a cell-based Human AhR Reporter Assay System, we demonstrated that melatonin and its indolic and kynuric metabolites act as agonists on the AhR with EC50's between 10-4 and 10-6 M. This was further validated via the stimulation of the transcriptional activation of the CYP1A1 promoter. Furthermore, melatonin and its metabolites stimulated AhR translocation from the cytoplasm to the nucleus in human keratinocytes, as demonstrated by ImageStream II cytometry and Western blot (WB) analyses of cytoplasmic and nuclear fractions of human keratinocytes. These functional analyses are supported by in silico analyses. We also investigated the peroxisome proliferator-activated receptor (PPAR)γ as a potential target for melatonin and metabolites bioregulation. The binding studies using a TR-TFRET kit to assay the interaction of the ligand with the ligand-binding domain (LBD) of the PPARγ showed agonistic activities of melatonin, 6-hydroxymelatonin and N-acetyl-N-formyl-5-methoxykynuramine with EC50's in the 10-4 M range showing significantly lower affinities that those of rosiglitazone, e.g., a 10-8 M range. These interactions were substantiated by stimulation of the luciferase activity of the construct containing PPARE by melatonin and its metabolites at 10-4 M. As confirmed by the functional assays, binding mode predictions using a homology model of the AhR and a crystal structure of the PPARγ suggest that melatonin and its metabolites, including 6-hydroxymelatonin, 5-methoxytryptamine and N-acetyl-N-formyl-5-methoxykynuramine, are excellent candidates to act on the AhR and PPARγ with docking scores comparable to their corresponding natural ligands. Melatonin and its metabolites were modeled into the same ligand-binding pockets (LBDs) as their natural ligands. Thus, functional assays supported by molecular modeling have shown that melatonin and its indolic and kynuric metabolites can act as agonists on the AhR and they can interact with the PPARγ at high concentrations. This provides a mechanistic explanation for previously reported cytoprotective actions of melatonin and its metabolites that require high local concentrations of the ligands to reduce cellular damage under elevated oxidative stress conditions. It also identifies these compounds as therapeutic agents to be used at pharmacological doses in the prevention or therapy of skin diseases.


Subject(s)
Melatonin , Receptors, Aryl Hydrocarbon , Humans , Keratinocytes/metabolism , Ligands , Melatonin/metabolism , PPAR gamma/metabolism , Receptors, Aryl Hydrocarbon/metabolism
2.
J Toxicol Environ Health B Crit Rev ; 24(8): 355-394, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34542016

ABSTRACT

In the wake of the Deepwater Horizon (DWH) oil spill, a number of government agencies, academic institutions, consultants, and nonprofit organizations conducted lab- and field-based research to understand the toxic effects of the oil. Lab testing was performed with a variety of fish, birds, turtles, and vertebrate cell lines (as well as invertebrates); field biologists conducted observations on fish, birds, turtles, and marine mammals; and epidemiologists carried out observational studies in humans. Eight years after the spill, scientists and resource managers held a workshop to summarize the similarities and differences in the effects of DWH oil on vertebrate taxa and to identify remaining gaps in our understanding of oil toxicity in wildlife and humans, building upon the cross-taxonomic synthesis initiated during the Natural Resource Damage Assessment. Across the studies, consistency was found in the types of toxic response observed in the different organisms. Impairment of stress responses and adrenal gland function, cardiotoxicity, immune system dysfunction, disruption of blood cells and their function, effects on locomotion, and oxidative damage were observed across taxa. This consistency suggests conservation in the mechanisms of action and disease pathogenesis. From a toxicological perspective, a logical progression of impacts was noted: from molecular and cellular effects that manifest as organ dysfunction, to systemic effects that compromise fitness, growth, reproductive potential, and survival. From a clinical perspective, adverse health effects from DWH oil spill exposure formed a suite of signs/symptomatic responses that at the highest doses/concentrations resulted in multi-organ system failure.


Subject(s)
Environmental Exposure/adverse effects , Petroleum Pollution/adverse effects , Water Pollutants, Chemical/toxicity , Animals , Birds , Environmental Monitoring/methods , Fishes , Humans , Multiple Organ Failure/etiology , Petroleum/toxicity , Turtles , Vertebrates
3.
J Pharmacol Exp Ther ; 378(2): 157-165, 2021 08.
Article in English | MEDLINE | ID: mdl-34074713

ABSTRACT

Viral-mediated in vivo gene delivery methods currently dominate among therapeutic strategies within the clinical and experimental settings, albeit with well documented limitations arising from immunologic constraints. In this study, we demonstrate the utility of nonviral hepatotropic in vivo gene delivery of unpackaged expression constructs, including one encoding fibroblast growth factor 21 (FGF21). FGF21 is an important hepatokine whose expression positively correlates with therapeutic outcomes across various animal models of obesity. Our data demonstrate that FGF21 expression can be restored into the livers of immunocompetent FGF21 knockout mice for at least 2 weeks after a single injection with an FGF21 expression plasmid. In wild-type C57BL6/J mice, in vivo transfection with an FGF21-expressing plasmid induced weight loss, decreased adiposity, and activated thermogenesis in white fat within 2 weeks. Furthermore, in vivo FGF21 gene delivery protected C57BL6/J mice against diet-induced obesity by decreasing adiposity and increasing uncoupling protein 1-dependent thermogenesis in brown fat and by boosting respiratory capacity in subcutaneous and perigonadal white fat. Together, the data illustrate a facile and effective methodology for delivering prolonged protein expression specifically to the liver. We contend that this method will find utility in basic science research as a practical means to enhance in vivo studies characterizing liver protein function. We further believe our data provide a rationale for further exploring the potential clinical utility of nonviral gene therapy in mouse models of disease. SIGNIFICANCE STATEMENT: This study presents a valuable method for nonviral gene delivery in mice that improves upon existing techniques. The data provide a rationale for further exploring the potential clinical utility of nonviral gene therapy in mouse models of disease and will likely enhance in vivo studies characterizing liver protein function.


Subject(s)
Fibroblast Growth Factors , Adipose Tissue, Brown , Animals , Mice , Protein Processing, Post-Translational
4.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374508

ABSTRACT

The aryl hydrocarbon receptor (AHR) has been studied for over 40 years, yet our understanding of this ligand-activated transcription factor remains incomplete. Each year, novel findings continually force us to rethink the role of the AHR in mammalian biology. The AHR has historically been studied within the context of potent activation via AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), with a focus on how the AHR mediates TCDD toxicity. Research has subsequently revealed that the AHR is actively involved in distinct physiological processes ranging from the development of the liver and reproductive organs, to immune system function and wound healing. More recently, the AHR was implicated in the regulation of energy metabolism and is currently being investigated as a potential therapeutic target for obesity. In this review, we re-trace the steps through which the early toxicological studies of TCDD led to the conceptual framework for the AHR as a potential therapeutic target in metabolic disease. We additionally discuss the key discoveries that have been made concerning the role of the AHR in energy metabolism, as well as the current and future directions of the field.


Subject(s)
Energy Metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Dioxins/adverse effects , Disease Models, Animal , Disease Susceptibility , Drug Development , Energy Metabolism/genetics , Gene Expression Regulation , Humans , Ligands , Mice, Transgenic , Molecular Targeted Therapy , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Polychlorinated Dibenzodioxins/adverse effects , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Wasting Syndrome/etiology , Wasting Syndrome/metabolism
5.
Int J Mol Sci ; 20(4)2019 Feb 22.
Article in English | MEDLINE | ID: mdl-30813227

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor highly expressed in hepatocytes. Researchers have employed global and liver-specific conditional Ahr knockout mouse models to characterize the physiological roles of the AHR; however, the gestational timing of AHR loss in these models can complicate efforts to distinguish the direct and indirect effects of post-gestational AHR deficiency. Utilizing a novel tamoxifen-inducible AHR knockout mouse model, we analyzed the effects of hepatocyte-targeted AHR loss in adult mice. The data demonstrate that AHR deficiency significantly reduces weight gain and adiposity, and increases multilocular lipid droplet formation within perigonadal white adipose tissue (gWAT). Protein and mRNA expression of fibroblast growth factor 21 (FGF21), an important hepatokine that activates thermogenesis in brown adipose tissue (BAT) and gWAT, significantly increases upon AHR loss and correlates with a significant increase of BAT and gWAT respiratory capacity. Confirming the role of FGF21 in mediating these effects, this phenotype is reversed in mice concomitantly lacking AHR and FGF21 expression. Chromatin immunoprecipitation analyses suggest that the AHR may constitutively suppress Fgf21 transcription through binding to a newly identified xenobiotic response element within the Fgf21 promoter. The data demonstrate an important AHR-FGF21 regulatory axis that influences adipose biology and may represent a "druggable" therapeutic target for obesity and its related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cell Respiration , Fibroblast Growth Factors/metabolism , Gonads/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Thermogenesis , Adipose Tissue, White/drug effects , Adiposity/drug effects , Animals , Body Weight/drug effects , Cell Respiration/drug effects , Diet, High-Fat , Drinking , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Fibroblast Growth Factors/genetics , Gonads/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Droplets/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , Phenotype , Physical Conditioning, Animal , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Tamoxifen/pharmacology , Thermogenesis/drug effects , Transcription, Genetic/drug effects
6.
Mol Pharmacol ; 92(3): 366-374, 2017 09.
Article in English | MEDLINE | ID: mdl-28696214

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates a plethora of target genes. Historically, the AhR has been studied as a regulator of xenobiotic metabolizing enzyme genes, notably cytochrome P4501A1 encoded by CYP1A1, in response to the exogenous prototypical ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). AhR activity depends on its binding to the xenobiotic response element (XRE) in partnership with the AhR nuclear translocator (Arnt). Recent studies identified stanniocalcin 2 (Stc2) as a novel AhR target gene responsive to the endogenous AhR agonist cinnabarinic acid (CA). CA-dependent AhR-XRE-mediated Stc2 upregulation is responsible for cytoprotection against ectoplasmic reticulum/oxidative stress-induced apoptosis both in vitro and in vivo. Significantly, CA but not TCDD induces expression of Stc2 in hepatocytes. In contrast to TCDD, CA is unable to induce the CYP1A1 gene, thus revealing an AhR agonist-specific mutually exclusive dichotomous transcriptional response. Studies reported here provide a mechanistic explanation for this differential response by identifying an interaction between the AhR and the metastasis-associated protein 2 (MTA2). Moreover, the AhR-MTA2 interaction is CA-dependent and results in MTA2 recruitment to the Stc2 promoter, concomitant with agonist-specific epigenetic modifications targeting histone H4 lysine acetylation. The results demonstrate that histone H4 acetylation is absolutely dependent on CA-induced AhR and MTA2 recruitment to the Stc2 regulatory region and induced Stc2 gene expression, which in turn confers cytoprotection to liver cells exposed to chemical insults.


Subject(s)
Epigenesis, Genetic , Glycoproteins/genetics , Oxazines/pharmacology , Receptors, Aryl Hydrocarbon/physiology , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Cytoprotection , Female , Histones/metabolism , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Polychlorinated Dibenzodioxins/pharmacology , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/agonists , Response Elements/physiology
7.
Curr Opin Toxicol ; 2: 87-92, 2017 Feb.
Article in English | MEDLINE | ID: mdl-32296737

ABSTRACT

Decades of research on the Aryl hydrocarbon Receptor (AhR) has unveiled its involvement in the toxicity of halogenated and polycyclic aromatic hydrocarbons, and a myriad of normal physiological processes. The molecular dissection of AhR biology has centered on a canonical signaling pathway in an effort to mechanistically reconcile the diverse pathophysiological effects of exposure to environmental pollutants. As a consequence, we now know that canonical signaling can explain many but not all of the AhR-mediated effects. Here we describe recent findings that point to non-canonical signaling pathways, and focus on a novel AhR interaction with the Krüppel-like Factor 6 protein responsible for previously un-recognized epigenetic changes in the chromatin affecting gene expression.

8.
PLoS One ; 11(10): e0165485, 2016.
Article in English | MEDLINE | ID: mdl-27798657

ABSTRACT

Acinar cells represent the primary target in necroinflammatory diseases of the pancreas, including pancreatitis. The signaling pathways guiding acinar cell repair and regeneration following injury remain poorly understood. The purpose of this study was to determine the importance of Hepatocyte Growth Factor Receptor/MET signaling as an intrinsic repair mechanism for acinar cells following acute damage and chronic alcohol-associated injury. Here, we generated mice with targeted deletion of MET in adult acinar cells (MET-/-). Acute and repetitive pancreatic injury was induced in MET-/- and control mice with cerulein, and chronic injury by feeding mice Lieber-DeCarli diets containing alcohol with or without enhancement of repetitive pancreatic injury. We examined the exocrine pancreas of these mice histologically for acinar death, edema, inflammation and collagen deposition and changes in the transcriptional program. We show that MET expression is relatively low in normal adult pancreas. However, MET levels were elevated in ductal and acinar cells in human pancreatitis specimens, consistent with a role for MET in an adaptive repair mechanism. We report that genetic deletion of MET in adult murine acinar cells was linked to increased acinar cell death, chronic inflammation and delayed recovery (regeneration) of pancreatic exocrine tissue. Notably, increased pancreatic collagen deposition was detected in MET knockout mice following repetitive injury as well alcohol-associated injury. Finally, we identified specific alterations of the pancreatic transcriptome associated with MET signaling during injury, involved in tissue repair, inflammation and endoplasmic reticulum stress. Together, these data demonstrate the importance of MET signaling for acinar repair and regeneration, a novel finding that could attenuate the symptomology of pancreatic injury.


Subject(s)
Acinar Cells/enzymology , Acinar Cells/pathology , Pancreas/enzymology , Pancreas/injuries , Proto-Oncogene Proteins c-met/metabolism , Wound Healing , Acute Disease , Alcohol Drinking/pathology , Animals , Ceruletide , Chronic Disease , Collagen/metabolism , Disease Models, Animal , Gene Deletion , Humans , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Pancreas/pathology , Pancreatitis, Chronic/enzymology , Pancreatitis, Chronic/pathology , Regeneration
9.
J Biol Chem ; 290(46): 27767-78, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26424795

ABSTRACT

The aryl hydrocarbon receptor (AhR), a regulator of xenobiotic toxicity, is a member of the eukaryotic Per-Arnt-Sim domain protein family of transcription factors. Recent evidence identified a novel AhR DNA recognition sequence called the nonconsensus xenobiotic response element (NC-XRE). AhR binding to the NC-XRE in response to activation by the canonical ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin resulted in concomitant recruitment of carbamoyl phosphate synthase 1 (CPS1) to the NC-XRE. Studies presented here demonstrate that CPS1 is a bona fide nuclear protein involved in homocitrullination (hcit), including a key lysine residue on histone H1 (H1K34hcit). H1K34hcit represents a hitherto unknown epigenetic mark implicated in enhanced gene expression of the peptidylarginine deiminase 2 gene, itself a chromatin-modifying protein. Collectively, our data suggest that AhR activation promotes CPS1 recruitment to DNA enhancer sites in the genome, resulting in a specific enzyme-independent post-translational modification of the linker histone H1 protein (H1K34hcit), pivotal in altering local chromatin structure and transcriptional activation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Citrulline/analogs & derivatives , Epigenesis, Genetic , Histones/metabolism , Nuclear Proteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Base Sequence , Cells, Cultured , Chromatin/metabolism , Chromatin/ultrastructure , Citrulline/metabolism , Female , Hydrolases/genetics , Lysine/metabolism , Mice , Mice, Inbred C57BL , Polychlorinated Dibenzodioxins/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein-Arginine Deiminases , Response Elements , Transcriptional Activation
10.
World J Hepatol ; 7(10): 1312-24, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26052377

ABSTRACT

Hepatocellular carcinoma (HCC)-related mortality is high because early detection modalities are hampered by inaccuracy, expense and inherent procedural risks. Thus there is an urgent need for minimally invasive, highly specific and sensitive biomarkers that enable early disease detection when therapeutic intervention remains practical. Successful therapeutic intervention is predicated on the ability to detect the cancer early. Similar unmet medical needs abound in most fields of medicine and require novel methodological approaches. Proteomic profiling of body fluids presents a sensitive diagnostic tool for early cancer detection. Here we describe such a strategy of comparative proteomics to identify potential serum-based biomarkers to distinguish high-risk chronic hepatitis C virus infected patients from HCC patients. In order to compensate for the extraordinary dynamic range in serum proteins, enrichment methods that compress the dynamic range without surrendering proteome complexity can help minimize the problems associated with many depletion methods. The enriched serum can be resolved using 2D-difference in-gel electrophoresis and the spots showing statistically significant changes selected for identification by liquid chromatography-tandem mass spectrometry. Subsequent quantitative verification and validation of these candidate biomarkers represent an obligatory and rate-limiting process that is greatly enabled by selected reaction monitoring (SRM). SRM is a tandem mass spectrometry method suitable for identification and quantitation of target peptides within complex mixtures independent on peptide-specific antibodies. Ultimately, multiplexed SRM and dynamic multiple reaction monitoring can be utilized for the simultaneous analysis of a biomarker panel derived from support vector machine learning approaches, which allows monitoring a specific disease state such as early HCC. Overall, this approach yields high probability biomarkers for clinical validation in large patient cohorts and represents a strategy extensible to many diseases.

11.
J Pharmacol Exp Ther ; 353(1): 201-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25672339

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a cytosolic ligand-activated transcription factor historically known for its role in xenobiotic metabolism. Although AhR activity has previously been shown to play a cytoprotective role against intrinsic apoptotic stimuli, the underlying mechanism by which AhR confers cytoprotection against apoptosis is largely unknown. Here, we demonstrate that activation of AhR by the tryptophan catabolite cinnabarinic acid (CA) directly upregulates expression of stanniocalcin 2 (Stc2) to elicit cytoprotection against apoptosis induced by endoplasmic reticulum stress and oxidative stress. Chromatin immunoprecipitation studies demonstrated that CA treatment induces direct AhR binding to a region of the Stc2 promoter containing multiple xenobiotic response elements. Using isolated primary hepatocytes from AhR wild-type (AhR floxed) and liver-specific AhR conditional knockout mice, we showed that pretreatment with CA conferred cytoprotection against hydrogen peroxide (H(2)O(2))-, thapsigargin-, and ethanol-induced apoptosis in an AhR-dependent manner. Furthermore, suppressing Stc2 expression using RNA interference confirmed that the cytoprotective properties of CA against H(2)O(2), thapsigargin, and ethanol injury were absolutely dependent on Stc2. Immunochemistry revealed the presence of Stc2 in the endoplasmic reticulum and on the cell surface, consistent with Stc2 secretion and autocrine and/or paracrine signaling. Finally, in vivo data using a mouse model of acute alcohol hepatotoxicity demonstrated that CA provided cytoprotection against ethanol-induced apoptosis, hepatic microvesicular steatosis, and liver injury. Collectively, our data uncovered a novel mechanism for AhR-mediated cytoprotection in the liver that is dependent on CA-induced Stc2 activity.


Subject(s)
Endoplasmic Reticulum Stress , Glycoproteins/biosynthesis , Liver/cytology , Oxazines/pharmacology , Oxidative Stress , Receptors, Aryl Hydrocarbon/metabolism , Animals , Apoptosis/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cytoprotection , Endoplasmic Reticulum/metabolism , Ethanol/pharmacology , Glycoproteins/genetics , Hydrogen Peroxide/pharmacology , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Liver/drug effects , Liver/metabolism , Mice, Knockout , Oxazines/metabolism , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/genetics , Thapsigargin/pharmacology , Up-Regulation
12.
Mol Pharmacol ; 85(4): 533-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24431146

ABSTRACT

Previous studies in hepatocyte-derived cell lines and the whole liver established that the aryl hydrocarbon receptor (AhR) can disrupt G1-phase cell cycle progression following exposure to persistent AhR agonists, such as TCDD (dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin). Growth arrest was attributed to inhibition of G1-phase cyclin-dependent kinase 2 (CDK2) activity. The present study examined the effect of TCDD exposure on liver regeneration following 70% partial hepatectomy in mice lacking the Cip/Kip inhibitors p21(Cip1) or p27(Kip1) responsible for regulating CDK2 activity. Assessment of the regenerative process in wild-type, p21(Cip1) knockout, and p27(Kip1) knockout mice confirmed that TCDD-induced inhibition of liver regeneration is entirely dependent on p21(Cip1) expression. Compared with wild-type mice, the absence of p21(Cip1) expression completely abrogated the TCDD inhibition, and accelerated hepatocyte progression through G1 phase during the regenerative process. Analysis of the transcriptional response determined that increased p21(Cip1) expression during liver regeneration involved an AhR-dependent mechanism. Chromatin immunoprecipitation studies revealed that p21(Cip1) induction required AhR binding to the newly characterized nonconsensus xenobiotic response element, in conjunction with the tumor suppressor protein Kruppel-like factor 6 functioning as an AhR binding partner. The evidence also suggests that AhR functionality following partial hepatectomy is dependent on a p21(Cip1)-regulated signaling process, intimately linking AhR biology to the G1-phase cell cycle program.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Liver Regeneration , Receptors, Aryl Hydrocarbon/metabolism , Response Elements , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hepatectomy , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polychlorinated Dibenzodioxins/toxicity , Proto-Oncogene Proteins/metabolism , Receptors, Aryl Hydrocarbon/agonists
13.
J Pharmacol Exp Ther ; 345(3): 419-29, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23512538

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-mediated basic helix-loop-helix transcription factor of the Per/Arnt/Sim family that regulates adaptive and toxic responses to a variety of chemical pollutants, including polycyclic aromatic hydrocarbons and halogenated aromatic hydrocarbons, most notably 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Ligand activation leads to AhR nuclear translocation and binding to a xenobiotic response element (XRE) in association with the Arnt to regulate gene expression. Several recent genome-wide transcriptional studies identified numerous AhR target genes that lack the canonical XRE recognition site in the promoter regions. Characterization of one such target gene, the plasminogen activator inhibitor 1, identified a novel nonconsensus XRE (NC-XRE) that confers TCDD responsiveness independently of the Arnt protein. Studies reported here show that the NC-XRE is a recognition site for the AhR and a new binding partner, the Kruppel-like factor (KLF) family member KLF6. In vivo chromatin immunoprecipitations and in vitro DNA binding studies demonstrate that the AhR and KLF6 proteins form an obligatory heterodimer necessary for NC-XRE binding. Mutational analyses show that the protein-protein interactions involve the AhR C terminus and KLF6 N terminus, respectively. Moreover, NC-XRE binding depends on the 5' basic region in KLF6 rather than the previously characterized zinc finger DNA binding domain. Collectively, the results unmask a novel AhR signaling mechanism distinct from the canonical XRE-driven process that will enrich our future understanding of AhR biology.


Subject(s)
DNA/metabolism , Kruppel-Like Transcription Factors/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Blotting, Western , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Environmental Pollutants , Female , Humans , Kruppel-Like Factor 6 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polychlorinated Dibenzodioxins/pharmacology , Promoter Regions, Genetic/drug effects , Protein Biosynthesis/drug effects , RNA-Binding Proteins/metabolism , Response Elements/genetics , Transcription, Genetic/drug effects , Xenobiotics/pharmacology , Zinc Fingers
14.
J Pharmacol Exp Ther ; 344(3): 579-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269473

ABSTRACT

Proper hepatocyte function is vital for survival; thus, unrepaired destruction of the parenchymal tissue leading to liver decompensation is devastating. Therefore, understanding the homeostatic process regulating liver regeneration is clinically important, and evidence that the aryl hydrocarbon receptor (AhR) can promote cell survival after intrinsic apoptotic stimuli is integral to the regenerative process. The current study uses primary hepatocytes to identify survival mechanisms consistent with normal AhR biology. Taking advantage of the Cre-lox system to manipulate AhR status, we designed a comprehensive microarray analysis to identify immediate and direct changes in the transcriptome concomitant with the loss of the AhR. As a result, we identified a unique data set with minimal overlap, compared with previous array studies, culminating in the identification of Stanniocalcin 2 (Stc2) as a novel receptor target gene previously reported to have a cytoprotective role in endoplasmic reticulum stress. The Stc2 promoter contains multiple putative xenobiotic response elements clustered in a 250-bp region that was shown to recruit the AhR by chromatin immunoprecipitation. Of interest, Stc2 gene expression is refractory to classic exogenous AhR agonists, but responds to cellular stress in an AhR-dependent mechanism consistent with a process promoting cell survival.


Subject(s)
Glycoproteins/metabolism , Hepatocytes/metabolism , Liver Regeneration/genetics , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Line , Cell Survival/genetics , Cytoprotection/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Female , Gene Expression , Glycoproteins/genetics , Hepatocytes/cytology , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Liver/cytology , Liver/metabolism , Liver/physiology , Mice , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/genetics , Response Elements , Transcriptome
15.
Mol Pharmacol ; 81(3): 338-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22113079

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a mediator of xenobiotic toxicity, best recognized for conveying the deleterious effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure. The AhR functions as a ligand-activated transcription factor that binds to a canonical xenobiotic response element (XRE) in association with the heterodimerization partner, the AhR nuclear translocator (Arnt) protein. However, within the repertoire of AhR target genes identified in recent years, many lack a clearly defined XRE highlighting the growing realization that AhR-mediated gene expression seems to involve additional mechanisms distinct from the well characterized process involving the XRE. The present study characterized a novel nonconsensus XRE (NC-XRE) in the promoter of the plasminogen activator inhibitor-1 (PAI-1) gene that recruits a novel protein-DNA complex responsible for TCDD-inducible expression. DNA binding studies and reporter assays identified key residues in the NC-XRE necessary for protein-DNA binding and function, respectively. Functional studies with AhR expression constructs confirm that TCDD-inducibility is AhR-dependent and requires direct AhR-DNA binding to the NC-XRE. Chromatin immunoprecipitation and RNA interference studies reveal that the Arnt protein is not a component of the NC-XRE-bound AhR complex, suggesting that in contrast to the XRE, AhR-dependent gene expression mediated through the NC-XRE may involve a new DNA binding partner.


Subject(s)
Gene Expression/physiology , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/physiology , Animals , Base Sequence , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , DNA Primers , Electrophoretic Mobility Shift Assay , Mice , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic
16.
Toxicology ; 276(2): 103-9, 2010 Oct 09.
Article in English | MEDLINE | ID: mdl-20637255

ABSTRACT

Mechanisms of hepatocyte proliferation triggered by tissue loss are distinguishable from those that promote proliferation in the intact liver in response to mitogens. Previous studies demonstrate that exogenous activation of the aryl hydrocarbon receptor (AhR), a soluble ligand-activated transcription factor in the basic helix-loop-helix family of proteins, suppresses compensatory liver regeneration elicited by surgical partial hepatectomy. The goal of the present study was to determine how AhR activation modulates hepatocyte cell cycle progression in the intact liver following treatment with the hepatomitogen, 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP). Mice were pretreated with the exogenous AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) 24h prior to treatment with TCPOBOP (3 mg/kg).). In contrast to the suppressive effects of AhR activation observed during compensatory regeneration, TCDD pretreatment resulted in a 30-50% increase in hepatocyte proliferation in the intact liver of TCPOBOP-treated mice. Although pretreatment with TCDD suppressed CDK2 kinase activity and increased the association of CDK2 with negative regulatory proteins p21Cip1 and p27Kip1, a corresponding increase in CDK4/cyclin D1 association and CDK4 activity which culminated in enhanced phosphorylation of retinoblastoma protein, consistent with the increased proliferative response. These findings are in stark contrast to previous observations that the activated AhR can suppress hepatocyte proliferation in vivo and reveal a new complexity to AhR-mediated cell cycle control.


Subject(s)
Hyperplasia/chemically induced , Liver/drug effects , Polychlorinated Dibenzodioxins/pharmacology , Pyridines/toxicity , Receptors, Aryl Hydrocarbon/drug effects , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/drug effects , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 4/metabolism , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Retinoblastoma Protein/drug effects , Retinoblastoma Protein/metabolism
17.
Biochem Pharmacol ; 77(6): 947-56, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19027718

ABSTRACT

The aryl hydrocarbon receptor (AhR) was implicated as a mediator of xenobiotic toxicity over three decades ago. Although a complete picture continues to elude us, investigations by many laboratories during the ensuing period have revealed much about AhR biology in normal physiological processes, as well as the toxicities induced by the dioxins and related polychlorinated aromatic hydrocarbons. The findings are captured in numerous excellent reviews. This commentary attempts to inject a new perspective on some new as well as frequently overlooked observations in the context of established receptor properties. Specifically, we examine the impact of transient versus sustained receptor activation on AhR biology, and explore the potential role for cytochrome P450 expression in regulating AhR activity amongst various tissues. The growing recognition that AhR action functions through multiple mechanisms serves to further highlight the importance of limiting prolonged receptor activation.


Subject(s)
Receptors, Aryl Hydrocarbon/physiology , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Aryl Hydrocarbon Hydroxylases/physiology , Humans , Organ Specificity/drug effects , Organ Specificity/physiology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/metabolism , Time Factors , Xenobiotics/metabolism , Xenobiotics/toxicity
18.
J Cell Biol ; 175(5): 709-14, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17130290

ABSTRACT

Fas receptor is a member of the tumor necrosis factor-alpha family of death receptors that mediate physiologic apoptotic signaling. To investigate the molecular mechanisms regulating calcium mobilization during Fas-mediated apoptosis, we have analyzed the sequential steps leading to altered calcium homeostasis and cell death in response to activation of the Fas receptor. We show that Fas-mediated apoptosis requires endoplasmic reticulum-mediated calcium release in a mechanism dependent on phospholipase C-gamma1 (PLC-gamma1) activation and Ca2+ release from inositol 1,4,5-trisphosphate receptor (IP3R) channels. The kinetics of Ca2+ release were biphasic, demonstrating a rapid elevation caused by PLC-gamma1 activation and a delayed and sustained increase caused by cytochrome c binding to IP3R. Blocking either phase of Ca2+ mobilization was cytoprotective, highlighting PLC-gamma1 and IP3R as possible therapeutic targets for disorders associated with Fas signaling.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Phospholipase C gamma/metabolism , fas Receptor/physiology , Apoptosis , Cell Line , Cytochromes c/metabolism , Cytochromes c/physiology , Fas Ligand Protein/metabolism , Fas Ligand Protein/physiology , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Models, Biological , Signal Transduction , fas Receptor/metabolism
19.
Cytometry A ; 69(7): 641-51, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16807893

ABSTRACT

BACKGROUND: Scanning cytometry now has many of the features (and power) of multiparameter flow cytometry while keeping its own advantages as an imaging technology. Modern instruments combine capabilities of scanning cytometry with the ability to manipulate cells. A new technology, called LEAP (laser-enabled analysis and processing), offers a unique combination of capabilities in cell purification and selective macromolecule delivery (optoinjection). METHODS: LEAP-mediated cell purification and optoinjection effects were assessed in model experiments using adherent and suspension cell types and cell mixtures plated and processed at different densities. Optoinjection effects were visualized by delivering fluorescent dextrans into cells. Results were analyzed using the LEAP instrument's own imaging system as well as by fluorescence and confocal microscopy. RESULTS: Live cell samples (adherent and suspension) could be purified to 90-100% purity with 50-90% yield, causing minimal cell damage depending on the cell type and plating density. Nearly one hundred percent of the targeted cells of all cell types examined could be successfully optoinjected with dextrans of 3-70 kDa, causing no visual damage to the cells. Indirect optoinjection effects were observed on untargeted cells within 5-60 microm to targeted areas under conditions used here. CONCLUSIONS: LEAP provides solutions in cell purification and targeted macromolecule delivery for traditional and challenging applications where other methods fall short.


Subject(s)
Hepatocytes/cytology , Laser Scanning Cytometry/methods , Animals , Cell Adhesion/physiology , Cell Separation/instrumentation , Cell Separation/methods , Cells, Cultured , HeLa Cells , Humans , Laser Scanning Cytometry/instrumentation , Mice , Mice, Inbred C57BL , Staining and Labeling
20.
Mol Pharmacol ; 70(1): 163-70, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16636136

ABSTRACT

In hepatocyte-derived cell lines, either loss of aryl hydrocarbon receptor (AhR) function or treatment with a persistent AhR agonist such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can disrupt G1 phase cell cycle progression. The present study used liver regeneration to explore mechanistically how AhR activity modulates hepatocyte proliferation in vivo. Treatment of mice with 20 mug/kg TCDD 1 day before 70% partial hepatectomy (PH) resulted in a 50 to 75% suppression in liver regeneration. Impaired proliferation was not associated with changes in levels of interleukin-6 or tumor necrosis factor-alpha, which prime quiescent hepatocytes to enter G1 phase. In fact, administration of TCDD 12 h after PH, a period well beyond the priming phase, still induced the G1 arrest. Decreased proliferation in TCDD-treated mice correlated with reduced cyclin-dependent kinase-2 (CDK2) activity, a pivotal regulator of G1/S phase transition. In contrast to observations made in cell culture, suppressed CDK2 activity was not strictly associated with increased binding of the CDK2 inhibitors p21Cip1 or p27Kip1. However, TCDD decreased levels of cyclin E binding to CDK2, despite normal cyclin E expression. The evidence also suggests that TCDD-induced hepatic growth arrest depends upon sustained AhR activity because transient AhR activation in response to endogenous queues failed to suppress the regenerative response. These findings establish a functional role for the AhR in regulating normal cell cycle control during liver regeneration.


Subject(s)
Liver Regeneration/physiology , Liver/physiopathology , Receptors, Aryl Hydrocarbon/physiology , Animals , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 2/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Environmental Pollutants/toxicity , Female , G1 Phase/drug effects , Gene Expression/drug effects , Hepatectomy , Immunoprecipitation , Interleukin-6/biosynthesis , Liver/drug effects , Liver/metabolism , Liver Regeneration/drug effects , Mice , Mice, Inbred C57BL , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/agonists , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...