Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Biochem Mol Toxicol ; 38(6): e23736, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38769691

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor. We previously reported spontaneous ileocecal tumorigenesis in AhR-deficient mice after the age of 10 weeks, which originated in the confined area between ileum and cecum. This study aimed to investigate the underlying mechanism that causes tumor development at this particular location. To observe mucosal architecture in detail, tissues of ileocecal region were stained with methylene blue. Gene expression profile in the ileocecal tissue was compared with cecum. Immunohistochemical analysis was performed with ileocecal tissues using antibodies against ileum-specific Reg3ß or cecum-specific Pitx2. In AhR+/+ mice and AhR+/- mice, that do not develop lesions, methylene blue staining revealed the gradually changing shape and arrangement of villi from ileum to cecum. It was also observed in AhR-deficient mice before developing lesions. Microarray-based analysis revealed abundant antimicrobial genes, such as Reg3, in the ileocecal tissue while FGFR2 and Pitx2 were specific to cecum. Immunohistochemical analysis of AhR-deficient mice indicated that lesions originated from the ileocecal junction, a boundary area between different epithelial types. Site-specific gene expression analysis revealed higher expression of IL-1ß at the ileocecal junction compared with the ileum or cecum of 9-11-week-old AhR-deficient mice. These findings indicate that AhR plays a vital function in the ileocecal junction. Regulating AhR activity can potentially manage the stability of ileocecal tissue possessing cancer-prone characteristics. This investigation contributes to understanding homeostasis in different epithelial transitional tissues, frequently associated with pathological states.


Subject(s)
Interleukin-1beta , Receptors, Aryl Hydrocarbon , Up-Regulation , Animals , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/deficiency , Mice , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Cecum/metabolism , Ileum/metabolism , Ileum/pathology , Mice, Knockout , Transcription Factors/genetics , Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors
2.
Biochem Biophys Rep ; 33: 101418, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36620088

ABSTRACT

Proteins are functionally regulated by various types of posttranslational modifications (PTMs). Ku, a heterodimer complex of Ku70 and Ku80 subunits, participates in DNA repair processes. Ku is distributed not only in the nucleus but also in the cytoplasm, suggesting that the function of Ku is regulated by its subcellular localization. Although Ku70 undergoes PTMs including phosphorylation or acetylation, it remains unknown whether the PTMs of Ku70 affect the subcellular localization of Ku. Using a cell-free pull-down assay technique, we show that Nε-acetylation of lysine residues in the synthetic peptide matched to Ku70's nuclear localization signal (NLS) reduces the peptide's interaction with the nuclear transport factor importin-α. The reduced interaction by acetylation was supported by molecular simulation analysis. In addition, when expressed in the endogenous Ku80-defective Chinese hamster ovary xrs-6 cells, some full-size human Ku70 mutants with substitutions of glutamine, a possible structural mimetic of Nε-acetyl-lysine, for lysine at the specific NLS positions exhibited no nuclear distribution. These findings imply that acetylation of particular lysine residues in the Ku70 NLS regulates nuclear localization of Ku.

3.
J Toxicol Sci ; 46(12): 611-618, 2021.
Article in English | MEDLINE | ID: mdl-34853246

ABSTRACT

The gastrointestinal tract is exposed to a myriad of mutagens, making the DNA damage response (DDR) essential to maintain intestinal homeostasis. In vivo models to study DDRs are necessary to understand the mechanisms of disease development caused by genetic disorders such as colorectal cancer. A double-stranded break (DSB) in DNA is the most toxic type of DNA damage; it can be induced by either X-rays or chemicals, including anticancer agents. If DSBs in DNA cannot be repaired, cells can die by apoptosis to be removed from tissues. Here, we show that the DDRs observed as the phosphorylation of H2AX (γH2AX) and caspase-3-dependent apoptosis-induction are under critical control in the intestine of C57BL mice that were injected intraperitoneally with bleomycin, a natural glycopeptide used clinically as an antitumor agent. We found a significant increase in γH2AX expression 2-6 hr post-treatment in mouse ileum, cecum, and colon tissues by Western blotting and immunostaining. Apoptotic cells were observed after 6-24 hr by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and immunofluorescence of active caspase-3. We observed that γH2AX expression and apoptotic cells were distributed in the lower part of the crypt. The experimental protocol described here is a simple procedure that can be used generally as an in vivo intestinal toxicity assay. Our experimental approach provides a useful method for examining the effects of various bioactive compounds on the DDR, which is essential for understanding intestinal homeostasis.


Subject(s)
Bleomycin , DNA Breaks, Double-Stranded , Animals , Apoptosis , Bleomycin/toxicity , DNA Damage , Histones/genetics , Mice , Mice, Inbred C57BL
4.
Exp Cell Res ; 343(2): 126-134, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26973338

ABSTRACT

Intestinal homeostasis is maintained by complex interactions between intestinal microorganisms and the gut immune system. Dysregulation of gut immunity may lead to inflammatory disorders and tumorigenesis. We previously have shown the tumor suppressive effects of aryl hydrocarbon receptor (AhR) in intestinal carcinogenesis. In the present study, we investigated AhR distribution in the mouse and human intestine by histochemical analysis. In the normal intestine, AhR was mainly localized in the stroma containing immune cells in the lamina propria and lymphoid follicles. On the other hand, in the tumor tissue from human colon cancer and that developed in Apc(Min/+)mice, AhR expression was elevated. AhR immunostaining was found in both stromal and tumor cells. Although AhR was localized in the cytoplasm of tumor cells in most cases, nuclear AhR was also observed in some. AhR knockdown using siRNA resulted in significant promotion of cell growth in colon cancer cell lines. Furthermore, AhR activation by AhR ligands supplemented in culture medium suppressed cell growth. Our study results suggest that tumor suppressive roles of AhR are estimated in two distinct ways: in normal tissue, AhR is associated with tumor prevention by regulating gut immunity, whereas in tumor cells, it is involved in growth suppression.


Subject(s)
Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , HCT116 Cells , HT29 Cells , Humans , Intestinal Neoplasms/pathology , Intestines/cytology , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/genetics , Rectal Neoplasms/metabolism , Rectal Neoplasms/pathology , Signal Transduction , Tissue Distribution
5.
Carcinogenesis ; 34(7): 1620-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23455376

ABSTRACT

The aryl hydrocarbon receptor (AhR) plays a suppressive role in cecal carcinogenesis by CUL4B/AhR-mediated ubiquitylation and degradation of ß-catenin, which is activated by xenobiotics and natural ligands. AhR-deficient (AhR(-)(/-)) mice develop cecal tumors with severe inflammation. To elucidate whether the tumors develop autonomously in AhR(-/-) mice due to impaired ß-catenin degradation or in association with accelerated inflammation, we performed two kinds of experiments using germ-free (GF) AhR(-/-) mice and compound mutant mice lacking genes for AhR and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), which plays an essential role in caspase-1 activation in inflammasomes. Both GF AhR(-/-) and AhR(-/-)•ASC(-/-) mice showed considerably reduced tumor development compared with that in AhR(-/-) mice albeit in a 'cancer-prone' state with aberrant ß-catenin accumulation. Blocking of the interleukin (IL)-1ß signaling pathway by treatment with a caspase-1 inhibitor, YVAD, reduced cecal tumorigenesis in AhR(-/-) mice. Signal transducers and activators of transcription 3 (STAT3) activation was detected in the cecal epithelium of the AhR(-/-) mice due to enhanced IL-6 production. An inhibitor of the STAT3 signaling pathway, AG490 suppressed the tumor formation. ASC-mediated inflammation was also found to play a critical role in tumor development in Apc(Min/+) mice, a mouse model of familial adenomatous polyposis. Collectively, these results revealed an important role of the bacteria-triggered or ASC-mediated inflammation signaling pathway in the intestinal tumorigenesis of mice and suggest a possible chemical therapeutic intervention, including AhR ligands and inhibitors of the inflammation pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CARD Signaling Adaptor Proteins/metabolism , Cecal Neoplasms/pathology , Inflammation/pathology , Receptors, Aryl Hydrocarbon/metabolism , Adenomatous Polyposis Coli/immunology , Adenomatous Polyposis Coli/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CARD Signaling Adaptor Proteins/genetics , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cecal Neoplasms/immunology , Cell Line , Enzyme Activation , Female , Germ-Free Life , Inflammasomes/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/immunology , Interleukin-6/immunology , Intestines/immunology , Intestines/microbiology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk , Receptors, Aryl Hydrocarbon/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology , beta Catenin/immunology , beta Catenin/metabolism
6.
J Dermatol Sci ; 58(3): 211-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20478695

ABSTRACT

BACKGROUND: The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor. When environmental pollutants, including chemical carcinogens, bind to AhR, the receptor translocates to nucleus and transcriptionally activates target genes including drug metabolizing enzymes such as P450s. Recent studies have shown that AhR mediates various responses, including cellular growth, differentiation, immune system and development. OBJECTIVE: In this study, we investigated the physiological function of AhR in skin. METHODS: Distribution of AhR in murine skin was examined by immunohistochemistry. Expression of a target gene which is transcriptionally activated by AhR is analysed by RT-PCR. RESULTS: We found that AhR co-localizes with the transcriptional repressor B lymphocyte maturation protein 1 (Blimp1) in sebaceous gland. In this report, we show that expression of Blimp1 is induced by treatment with AhR ligands, such as methylcolanthrene (MC) in sebocyte and keratinocyte cell lines. Exposure to ultraviolet B, which has been reported to generate AhR ligand intracellularly, also increased Blimp1 mRNA. This ligand-dependent induction of Blimp1 requires the expression of both AhR and ARNT, since transfection of siRNA specific to either AhR or ARNT significantly reduced Blimp1 mRNA in response to MC. Analysis using kinase inhibitors revealed that ligand-dependent induction of Blimp1, but not that of CYP1A1, is inhibited by staurosporine. TPA, a potent activator of protein kinase C, increased Blimp1 mRNA but not CYP1A1. CONCLUSION: These data indicate that Blimp1 is a novel AhR-target gene in epidermal keratinocyte and sebocyte.


Subject(s)
Keratinocytes/physiology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Sebaceous Glands/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , B-Lymphocytes/physiology , Cell Line , Enzyme Inhibitors/pharmacology , Epidermal Cells , Epidermis/physiology , Humans , Immunohistochemistry , Keratinocytes/cytology , Keratinocytes/drug effects , Mice , Mice, Mutant Strains , Positive Regulatory Domain I-Binding Factor 1 , RNA, Messenger/metabolism , RNA, Small Interfering , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sebaceous Glands/cytology , Staurosporine/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
7.
Mol Cell Biol ; 29(24): 6391-400, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19822660

ABSTRACT

Aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, is known to mediate a wide variety of pharmacological and toxicological effects caused by polycyclic aromatic hydrocarbons. Recent studies have revealed that AhR is involved in the normal development and homeostasis of many organs. Here, we demonstrate that AhR knockout (AhR KO) mice are hypersensitive to lipopolysaccharide (LPS)-induced septic shock, mainly due to the dysfunction of their macrophages. In response to LPS, bone marrow-derived macrophages (BMDM) of AhR KO mice secreted an enhanced amount of interleukin-1beta (IL-1beta). Since the enhanced IL-1beta secretion was suppressed by supplementing Plasminogen activator inhibitor-2 (Pai-2) expression through transduction with Pai-2-expressing adenoviruses, reduced Pai-2 expression could be a cause of the increased IL-1beta secretion by AhR KO mouse BMDM. Analysis of gene expression revealed that AhR directly regulates the expression of Pai-2 through a mechanism involving NF-kappaB but not AhR nuclear translocator (Arnt), in an LPS-dependent manner. Together with the result that administration of the AhR ligand 3-methylcholanthrene partially protected mice with wild-type AhR from endotoxin-induced death, these results raise the possibility that an appropriate AhR ligand may be useful for treating patients with inflammatory disorders.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Receptors, Aryl Hydrocarbon/metabolism , Shock, Septic/chemically induced , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Basic Helix-Loop-Helix Transcription Factors , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Cytokines/immunology , Gene Expression Profiling , Humans , Interleukin-1beta/immunology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Plasminogen Activator Inhibitor 2/genetics , Plasminogen Activator Inhibitor 2/metabolism , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/genetics , Shock, Septic/immunology
8.
Proc Natl Acad Sci U S A ; 106(32): 13481-6, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19651607

ABSTRACT

Intestinal cancer is one of the most common human cancers. Aberrant activation of the canonical Wnt signaling cascade, for example, caused by adenomatous polyposis coli (APC) gene mutations, leads to increased stabilization and accumulation of beta-catenin, resulting in initiation of intestinal carcinogenesis. The aryl hydrocarbon receptor (AhR) has dual roles in regulating intracellular protein levels both as a ligand-activated transcription factor and as a ligand-dependent E3 ubiquitin ligase. Here, we show that the AhR E3 ubiquitin ligase has a role in suppression of intestinal carcinogenesis by a previously undescribed ligand-dependent beta-catenin degradation pathway that is independent of and parallel to the APC system. This function of AhR is activated by both xenobiotics and natural AhR ligands, such as indole derivatives that are converted from dietary tryptophan and glucosinolates by intestinal microbes, and suppresses intestinal tumor development in Apc(Min/+) mice. These findings suggest that chemoprevention with naturally-occurring and chemically-designed AhR ligands can be used to successfully prevent intestinal cancers.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Intestines/pathology , Precancerous Conditions/pathology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cecal Neoplasms/metabolism , Cecal Neoplasms/pathology , Intestinal Mucosa/metabolism , Ligands , Mice , Mice, Inbred C57BL , Precancerous Conditions/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Receptors, Aryl Hydrocarbon/deficiency , Signal Transduction , Ubiquitination , beta Catenin/metabolism
9.
Biochem Pharmacol ; 77(4): 588-96, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18983832

ABSTRACT

Because aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor, its nuclear translocation in response to ligands may be directly linked to transcriptional activation of target genes. We have investigated the biological significance of AhR from the perspective of its subcellular localization and revealed that AhR possesses a functional nuclear localization signal (NLS) as well as a nuclear export signal (NES) which controls the distribution of AhR between the cytoplasm and nucleus. The intracellular localization of AhR is regulated by phosphorylation of amino acid residues in the vicinity of the NLS and NES. In cell culture systems, cell density affects not only its intracellular distribution of AhR, but also its transactivation activity of the target genes such as transcriptional repressor Slug, which is important for the induction of epithelial-mesenchymal transitions. These effects of AhR observed in cultured cells are proposed to be reflected on the in vivo response such as morphogenesis and tumor formation. This review summarizes recent work on the control mechanism of AhR localization and progress in understanding the physiological role of AhR in the skin. We propose that AhR is involved in normal skin formation during fetal development as well as in pathological states such as epidermal wound healing and skin carcinogenesis.


Subject(s)
Receptors, Aryl Hydrocarbon/metabolism , Skin Neoplasms/metabolism , Skin Physiological Phenomena , Skin/metabolism , Animals , Cell Count , Humans , Polychlorinated Dibenzodioxins/toxicity , Protein Transport , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/physiology , Skin/injuries , Skin/pathology , Skin Neoplasms/pathology , Skin Physiological Phenomena/drug effects , Wound Healing/drug effects , Wound Healing/physiology
10.
Exp Cell Res ; 312(18): 3585-94, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16952353

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor. We previously showed that AhR localizes predominantly in the cytoplasm under high cell densities of a keratinocytes cell line, HaCaT, but accumulates in the nucleus at low cell densities. In the current report, we show that the Slug, which is a member of the snail/slug family of zinc finger transcriptional repressors critical for induction of epithelial-mesenchymal transitions (EMT), is activated transcriptionally in accordance with nuclear accumulation of AhR. By reporter assay of the promoter of the Slug gene, gel shift and chromatin immunoprecipitation analyses showed AhR directly binds to xenobiotic responsive element 5 at -0.7 kb of the gene. AhR-targeted gene silencing by small interfering RNA duplexes led to the abolishment of not only CYP1A1 but also Slug induction by 3-methycholanthrene. The Slug was co-localized to the AhR at the wound margins of HaCaT cells, where apparent nuclear distribution of AhR and Slug was observed. The induced Slug was associated with reduction of an epithelial marker of cytokeratin-18 and with an increase in the mesenchymal marker, fibronectin. Taken together, these findings suggest that AhR participated in Slug induction, which, in turn, regulates cellular physiology including cell adhesion and migration.


Subject(s)
Gene Expression Regulation , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Zinc Fingers , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Genes, Reporter , Humans , Mice , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics
11.
Int J Oncol ; 29(3): 689-93, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16865286

ABSTRACT

A t(11;22)(q24;q12) translocation is present in 90% of Ewing's sarcoma, and results in the formation of the EWS-FLI1 fusion gene encoding an oncogenic transcription factor. To clarify the function of chimeric EWS-FLI1 proteins, an identification of a nuclear localization signal (NLS) in the EWS, FLI1 and EWS-FLI1 proteins is important because the chimeric oncoprotein may lose or gain NLS function different from native proteins resulting in different subcellular localization, and in deregulated gene expression. Furthermore, some studies reported that patients with one type of fusion gene ('type 1') had better overall survival than those with other types, suggesting that functional differences may be present among various fusion proteins. There has been only one study reporting a NLS in EWS, but none reporting those in FLI1 and EWS-FLI1. To clarify the molecular mechanisms of Ewing tumor development, we first identified the NLSs of EWS and FLI1. We allocated the NLS to amino acid residues 632-656 near the C-terminal region of EWS that is different from the previous study, and identified two NLSs of FLI1, NLS1 (63-90) in the N-terminal domain and NLS2 (319-360) in the 3'-ETS domain. In addition, the present study showed that all of the EWS-FLI1 fusion proteins completely reside in the nucleus without affecting the frequency of nuclear localization among variants, suggesting that NLS2 of FLI1 was used for nuclear translocation of various EWS-FLI1 fusion proteins.


Subject(s)
Bone Neoplasms/metabolism , Cell Nucleus/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Transport , Proto-Oncogene Protein c-fli-1/metabolism , Sarcoma, Ewing/metabolism , Transcription Factors/physiology , Blotting, Western , Fluorescent Antibody Technique, Indirect , Green Fluorescent Proteins/metabolism , HeLa Cells/metabolism , Humans , Nuclear Proteins , Plasmids , Protein Structure, Tertiary , RNA-Binding Protein EWS , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
12.
Biochem Biophys Res Commun ; 331(4): 902-8, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15882963

ABSTRACT

Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin induces developmental toxicity in reproductive organs. To elucidate the function of AhR, we generated stable transformants of TM3 cells overexpressing wild-type aryl hydrocarbon receptor (AhR) or its mutants which carried mutations in nuclear localization signal or nuclear export signal. In the presence of 3-methylcholanthrene (MC), proliferation of the cells transfected with wild-type AhR was completely suppressed, whereas cells expressing AhR mutants proliferated in a manner equivalent to control TM3 cells, suggesting AhR-dependent growth inhibition. The suppression was associated with up-regulation of cyclin-dependent kinase inhibitor p21Cip1, which was abolished by pretreatment with actinomycin D. A p38 MAPK specific inhibitor, SB203580, blocked the increase of p21Cip1 mRNA in response to MC. Treatment with indigo, another AhR ligand, failed to increase of p21Cip1 mRNA, although up-regulation of mRNA for CYP1A1 was observed. These data suggest AhR in Leydig cells mediates growth inhibition by inducing p21Cip1.


Subject(s)
Cell Division/physiology , Leydig Cells/cytology , Receptors, Aryl Hydrocarbon/physiology , Animals , Base Sequence , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , DNA Primers , Leydig Cells/enzymology , Male , Mice , Mutagenesis, Site-Directed , Receptors, Aryl Hydrocarbon/genetics
13.
Biochem Biophys Res Commun ; 317(2): 545-50, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-15063792

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor which plays a role as an intracellular mediator of the xenobiotic signaling pathway. We previously identified the minimum nuclear localization signal (NLS) of AhR(13-39): it is composed of two basic amino acid segments, AhR(13-16:RKRR) and AhR(37-39:KRH). In this study, we showed that the two protein kinase C (PKC) sites of Ser-12 and Ser-36 are located one amino acid upstream from each of the two segments, and that a ligand-dependent nuclear import of AhR is inhibited by substitution of aspartic acid for Ser-12 (S12D) or Ser-36 (S36D), which mimics the negative charge of phosphorylation. This observation was supported by microinjection analysis, an in vitro nuclear transport assay, and a luciferase reporter assay, suggesting a two-step mechanism in the ligand-dependent nuclear translocation of AhR.


Subject(s)
Cell Nucleus/metabolism , Liver/metabolism , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/physiology , Active Transport, Cell Nucleus/physiology , Animals , Binding Sites , Cell Line , Dogs , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Structure-Activity Relationship
14.
J Biol Chem ; 279(18): 19209-16, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14985336

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that plays a role as an intracellular mediator of the xenobiotic signaling pathway. AhR contains signals for both nuclear localization and nuclear export (NES). The objective of this study was to demonstrate how AhR intracellular distribution was regulated physiologically in cells. We found that cell density, but not the cell cycle, influenced the subcellular distribution of AhR in a keratinocyte cell line, HaCaT: AhR was predominantly nuclear at sparse cell densities, both nuclear and cytoplasmic at subconfluence, and predominantly cytoplasmic at confluence. Stable transfectants of HaCaT carrying a reporter gene fused with xenobiotic responsive element showed an association between xenobiotic responsive element-mediated transcription and AhR relocalization. Leptomycin B promoted nuclear accumulation of AhR irrespective of cell density, suggesting that this alteration may be because of a change of the regulation of the nuclear export of AhR. We found that Ser-68 in the NES of AhR was phosphorylated after nuclear accumulation of activated AhR and the nuclear export of a chimeric GST-AhR-GFP fusion protein was suppressed by substitution of a serine residue (Ser-68) to aspartic acid, which mimics the negative charge of phosphorylation. This novel cell density-dependent AhR relocalization was affected by exposure to SB203580, okadaic acid, and low Ca(2+) concentrations. These findings strongly suggest that cell density regulates the intracellular localization and function of AhR, because of modulation of nuclear export activity. The p38 MAPK-mediated phosphorylation of the NES and its dephosphorylation, regulated by cell-cell contact signals, may have pivotal roles in the novel AhR relocalization.


Subject(s)
Active Transport, Cell Nucleus , DNA-Binding Proteins , Keratinocytes/cytology , Receptors, Aryl Hydrocarbon/metabolism , Transcriptional Activation , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Cell Compartmentation , Cell Count , Cell Cycle , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Sorting Signals , Protein Transport , Receptors, Aryl Hydrocarbon/biosynthesis , Transcription Factors , Wound Healing , p38 Mitogen-Activated Protein Kinases
15.
Mol Endocrinol ; 17(6): 994-1004, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12610109

ABSTRACT

Dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (Dax-1, NR0B1) is an orphan nuclear receptor that represses transcription by Ad4 binding protein/steroidogenic factor 1 (Ad4BP/SF-1, NR5A1). Observations on human diseases and the phenotypes of mice, in which the corresponding genes have been disrupted, have elucidated essential roles of these two nuclear receptors in differentiation of steroidogenic tissues. However, little is known about how the functions of these factors are regulated. Here we have examined their subcellular localization and have clarified the molecular mechanisms regulating subcellular localization of Dax-1. Prompted by the finding that nuclear localization of Dax-1 correlates with the presence of Ad4BP/SF-1 in the early stages of pituitary development, we have tested the possibility that interaction between the two factors is essential for the nuclear localization of Dax-1. In vitro studies with cultured cells demonstrated that an interaction involving the LXXLL motifs in the N-terminal repeat region of Dax-1 plays a key role in its subcellular localization. In addition, we found that a mutant form of DAX-1 (L466R), from a patient with adrenal hypoplasia congenita, was defective in nuclear localization in spite of having an intact N terminus. Taken together, the results reveal that the subcellular localization of Dax-1 is influenced by the presence of Ad4BP/SF-1, and that two regions of Dax-1 have important roles for this process.


Subject(s)
Amino Acid Motifs/physiology , DNA-Binding Proteins/metabolism , Protein Structure, Tertiary/physiology , Receptors, Retinoic Acid/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cell Nucleus/physiology , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/genetics , Fushi Tarazu Transcription Factors , Gene Expression Regulation, Developmental/physiology , HeLa Cells , Homeodomain Proteins , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Pituitary Gland/embryology , Pituitary Gland/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, Retinoic Acid/genetics , Repressor Proteins/genetics , Steroidogenic Factor 1 , Structure-Activity Relationship , Subcellular Fractions/physiology , Transcription Factors/genetics
16.
Mol Pharmacol ; 63(3): 524-31, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12606758

ABSTRACT

The steroid and xenobiotic receptor (SXR) is an orphan nuclear receptor that plays a key role in the regulation of xenobiotic response by controlling the expression of drug metabolizing and clearance enzymes. We observed that pregnane X receptor (PXR), the mouse ortholog of SXR, was retained in the cytoplasm of hepatic cells of untreated mice, whereas PXR was translocated to the nucleus after administration of a ligand, pregnenolone 16 alpha-carbonitrile. To understand the molecular mechanisms underlying the xenochemical-dependent nuclear translocation of SXR, we identified the signal sequence of SXR that regulates its nuclear translocation; using an in vitro expression system, we allocated the nuclear localization signal (NLS) to amino acid residues 66 to 92 within the DNA binding domain of SXR. The NLS of SXR is characterized as the bipartite type, and is recognized by the three molecular species of importin alpha: Rch1 (PTAC58), NPI1, and Qip1, in the presence of PTAC97 of importin beta to target the nuclear pore. The nuclear translocation of SXR was observed as an essential regulatory event for transcription of its target genes such as CYP3A4. These results strongly suggest that the molecular mechanism of the nuclear import of SXR was different from that of another xenosensor, the constitutively active receptor, whose translocation into the nucleus is mediated by a leucine-rich xenochemical response signal in its ligand binding domain.


Subject(s)
Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Active Transport, Cell Nucleus , Animals , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Microinjections , Peptide Fragments/metabolism , Pregnane X Receptor , Recombinant Fusion Proteins/pharmacology , Subcellular Fractions , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured , alpha Karyopherins/metabolism
17.
J Biochem ; 131(1): 79-85, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11754738

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that acts in concert with the AhR nuclear translocator (ARNT). Subcellular localization and transcriptional activation of target genes are mainly regulated by ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We have previously reported that AhR migrates in cells as a nucleocytoplasmic shuttling protein mediated by its nuclear localization and export signals. A short sequence motif LxxLL (L is leucine and x is any amino acid) found in transcriptional co-activators has been reported to mediate the binding to liganded nuclear receptors. The role of the two LxxLL motifs, AhR[50-54] and [224-228], has now been analyzed by determining the localization of AhR and its transcriptional activity with Leu to Ala mutations in full-length AhR. Immunocytostaining revealed that mutation of the motif at AhR[50-54] promotes the efficiency of nuclear localization in the absence of ligand without altering HSP90 and ARA9 binding or nuclear export activity. Furthermore, this mutation decreases the transcriptional activity of the AhR/ARNT system, which is likely due to the suppression of AhR/ARNT/XRE complex formation. Another LxxLL motif at AhR[224-227] affects neither the subcellular localization nor transcriptional activity. These results indicate that the LxxLL motif at AhR[50-54] is important for the regulation of AhR activity.


Subject(s)
Receptors, Aryl Hydrocarbon/genetics , Transcriptional Activation , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , COS Cells/metabolism , Chlorocebus aethiops , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Leucine/chemistry , Leucine/genetics , Leucine/physiology , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/metabolism , Subcellular Fractions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...