Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biomedicines ; 10(8)2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35892692

ABSTRACT

Claudin proteins are intercellular adhesion molecules. Increased claudin domain-containing 1 (CLDND1) expression is associated with the malignant transformation of estrogen receptor-negative breast cancer cells with low sensitivity to hormone therapy. Abnormal CLDND1 expression is also implicated in vascular diseases. Previously, we investigated the regulatory mechanism underlying CLDND1 expression and identified a strong enhancer region near the promoter. In silico analysis of the sequence showed high homology to the ETS domain-containing protein-1 (ELK1)-binding sequence which is involved in cell growth, differentiation, angiogenesis, and cancer. Transcriptional ELK1 activation is associated with the mitogen-activated protein kinase (MAPK) signaling cascade originating from the epidermal growth factor receptor (EGFR). Here, we evaluated the effect of gefitinib, an EGFR tyrosine kinase inhibitor, on the suppression of CLDND1 expression using ELK1 overexpression in luciferase reporter and chromatin immunoprecipitation assays. ELK1 was found to be an activator of the enhancer region, and its transient expression increased that of CLDND1 at the mRNA and protein levels. CLDND1 expression was increased following EGF-induced ELK1 phosphorylation. Furthermore, this increase in CLDND1 was significantly suppressed by gefitinib. Therefore, EGF-dependent activation of ELK1 contributes to the induction of CLDND1 expression. These findings open avenues for the development of new anticancer agents targeting CLDND1.

2.
Pharmacol Rep ; 74(3): 503-512, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35366749

ABSTRACT

BACKGROUND: Lipoprotein metabolism is essential for the growth and proliferation of cancer cells, and is involved in the supply of energy and cellular components. Lipoprotein lipase (LPL) is a very important enzyme in lipoprotein metabolism; however, the details underlying the mechanism of LPL secretion are unclear. Palbociclib is an antitumor drug that inhibits cell cycle progression and suppresses the growth of cancer cells. The effects of palbociclib on energy metabolism, particularly on lipid metabolism, have not been fully elucidated. METHODS: We examined the regulation of LPL secretion, which is primarily involved in lipoprotein metabolism. FM3A mouse mammary tumor cells, which are hormone receptor-positive breast cancer cells, were treated with palbociclib, and the activity and protein levels of secreted LPL were measured. Moreover, the changes in intracellular lipid content were measured by fluorescence staining using Nile Red. RESULTS: FM3A cells were treated with palbociclib, the activity and protein content of secreted LPL were increased. The stimulatory secretion of LPL by palbociclib was suppressed by an intracellular Ca2+ chelator (BAPTA-AM) and a Ca2+/calmodulin-dependent protein kinase kinase (CaMKK) inhibitor (STO-609). Furthermore, the palbociclib-stimulated secretion of LPL was not observed in AMP-activated protein kinase (AMPK)-knockdown cells. An increase in the fluorescence intensity of Nile Red was observed in palbociclib-treated cells; however, no increase was observed in LPL-knockdown cells. CONCLUSIONS: Our data suggest that palbociclib causes intracellular lipid accumulation in breast cancer cells by stimulating Ca2+/CaMKK/AMPK-mediated LPL secretion.


Subject(s)
Breast Neoplasms , Lipoprotein Lipase , AMP-Activated Protein Kinases , Animals , Breast Neoplasms/drug therapy , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Female , Humans , Lipids , Lipoprotein Lipase/metabolism , Lipoproteins , Mice , Piperazines , Pyridines
3.
J Toxicol Sci ; 47(4): 159-168, 2022.
Article in English | MEDLINE | ID: mdl-35370244

ABSTRACT

Detailed in vitro studies on the effects of perfluorooctanoic acid (PFOA) have demonstrated that activation of peroxisome proliferator-activated receptor α (PPARα) is a key process by which PFOA affects the malignancy of estrogen receptor α (ERα)-positive breast cancer cells. However, there is very little information on the PPARα-regulated genes responsible for the effects of PFOA in ERα-negative breast cancer cell malignancy. We recently demonstrated that fatty acid 2-hydroxylase (FA2H) stimulates the migration of ERα-negative human MDA-MB-231 cells, and PPARα is a key factor for the induction of FA2H in these cells. However, evidence for the relationship between PFOA exposure and PPARα-FA2H axis-driven migration has not been obtained. Here we analyzed the effects of PFOA on PPARα transcription and FA2H expression in relation to MDA-MB-231 cell migration. We found that simultaneously with stimulated migration, PFOA upregulated FA2H and activated the transcription of PPARα. FA2H-selective siRNA, but not siRNA control, clearly dampened PFOA-mediated cell migration. There is an inhibitory interaction between PPARα and PPARß/δ (i.e., PPARß/δ can suppress PPARα-mediated transcription) in MDA-MB-231 cells, but even in the presence of PPARß/δ expression, PFOA appeared to free PPARα to upregulate FA2H. Collectively, our findings show that i) PFOA activates PPARα-mediated transcription, ii) PFOA stimulates migration dependent on FA2H expression, and iii) mechanistically, PFOA relieves PPARß/δ suppression of PPARα activity to upregulate FA2H in MDA-MB-231 cells.


Subject(s)
Receptors, Estrogen , Triple Negative Breast Neoplasms , Caprylates/toxicity , Cell Movement , Fluorocarbons , Humans , Mixed Function Oxygenases/genetics
4.
Biol Pharm Bull ; 43(9): 1407-1412, 2020.
Article in English | MEDLINE | ID: mdl-32879215

ABSTRACT

The role of ß-estradiol (E2) in lipoprotein metabolism in mammary tumors is unclear, therefore, we investigated the effect of E2 on the secretion of lipoprotein lipase (LPL) from mouse mammary tumor FM3A cells. E2-treated cells increased the secretion of active LPL from FM3A cells in a time- and dose-dependent manner. Activity of mitogen-activated protein kinase (MAPK) was increased in the tumor cells treated with E2, and enhanced secretion of LPL was suppressed by MAPK kinase 1/2 inhibitor, PD98059, extracellular signal-regulated kinase (ERK) 1/2 inhibitor, FR180204, p38 MAPK inhibitor, SB202190, and phosphatidyl inositol 3-kinase (PI3K) inhibitor, LY294002. In addition, the effect of E2 on LPL secretion was markedly suppressed by an inhibitor of mammalian target of rapamycin complex (mTORC) 1 and 2, KU0063794, but were not by a mTORC1 inhibitor, rapamycin. Furthermore, a small interfering RNA (siRNA)-mediated decrease in the expression of rapamycin-insensitive companion of mTOR (Rictor), a pivotal component of mTORC2, suppressed secretion of LPL by E2. These results suggest that the stimulatory secretion of LPL by E2 from the tumor cells is closely associated with an activation of mTORC2 rather than mTORC1 possibly via the MAPK cascade.


Subject(s)
Estradiol/metabolism , Lipoprotein Lipase/metabolism , MAP Kinase Signaling System/physiology , Mammary Neoplasms, Animal/pathology , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Animals , Cell Line, Tumor , Culture Media/metabolism , Female , Gene Knockdown Techniques , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipoproteins/metabolism , MAP Kinase Signaling System/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Rapamycin-Insensitive Companion of mTOR Protein/antagonists & inhibitors , Rapamycin-Insensitive Companion of mTOR Protein/genetics
5.
BMC Mol Cell Biol ; 21(1): 32, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32321446

ABSTRACT

BACKGROUND: Neutral cholesterol ester hydrolase 1 (NCEH1) catalyzes the hydrolysis of cholesterol ester (CE) in macrophages. Genetic ablation of NCEH1 promotes CE-laden macrophages and the development of atherosclerosis in mice. Dysregulation of NCEH1 levels is involved in the pathogenesis of multiple disorders including metabolic diseases and atherosclerosis; however, relatively little is known regarding the mechanisms regulating NCEH1. Retinoic acid receptor-related orphan receptor α (RORα)-deficient mice exhibit several phenotypes indicative of aberrant lipid metabolism, including dyslipidemia and increased susceptibility to atherosclerosis. RESULTS: In this study, inhibition of lipid droplet formation by RORα positively regulated NCEH1 expression in macrophages. In mammals, the NCEH1 promoter region was found to harbor putative RORα response elements (ROREs). Electrophoretic mobility shift, chromatin immunoprecipitation, and luciferase reporter assays showed that RORα binds and responds to ROREs in human NCEH1. Moreover, NCEH1 was upregulated through RORα via a phorbol myristate acetate-dependent mechanism during macrophage differentiation from THP1 cells. siRNA-mediated knockdown of RORα significantly downregulated NCEH1 expression and accumulated lipid droplets in human hepatoma cells. In contrast, NCEH1 expression and removal of lipid droplets were induced by RORα agonist treatments and RORα overexpression in macrophages. CONCLUSION: These data strongly suggested that NCEH1 is a direct RORα target, defining potential new roles for RORα in the inhibition of lipid droplet formation through NCEH1.


Subject(s)
Lipid Droplets/metabolism , Macrophages/metabolism , Receptors, Retinoic Acid/metabolism , Sterol Esterase/metabolism , Animals , Atherosclerosis/enzymology , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Line, Tumor , Cholesterol Esters/metabolism , Cholesterol, LDL/pharmacology , Chromatin Immunoprecipitation , Gene Knockdown Techniques , Humans , Macrophages/enzymology , Mice , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/genetics , Sterol Esterase/genetics , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation
6.
J Immunol ; 203(12): 3386-3394, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31694909

ABSTRACT

Human leukocyte Ig-like receptors (LILR) LILRB1 and LILRB2 are immune checkpoint receptors that regulate a wide range of physiological responses by binding to diverse ligands, including HLA-G. HLA-G is exclusively expressed in the placenta, some immunoregulatory cells, and tumors and has several unique isoforms. However, the recognition of HLA-G isoforms by LILRs is poorly understood. In this study, we characterized LILR binding to the ß2-microglobulin (ß2m)-free HLA-G1 isoform, which is synthesized by placental trophoblast cells and tends to dimerize and multimerize. The multimerized ß2m-free HLA-G1 dimer lacked detectable affinity for LILRB1, but bound strongly to LILRB2. We also determined the crystal structure of the LILRB1 and HLA-G1 complex, which adopted the typical structure of a classical HLA class I complex. LILRB1 exhibits flexible binding modes with the α3 domain, but maintains tight contacts with ß2m, thus accounting for ß2m-dependent binding. Notably, both LILRB1 and B2 are oriented at suitable angles to permit efficient signaling upon complex formation with HLA-G1 dimers. These structural and functional features of ligand recognition by LILRs provide novel insights into their important roles in the biological regulations.


Subject(s)
HLA-G Antigens/chemistry , Models, Molecular , Protein Conformation , Receptors, Immunologic/chemistry , Binding Sites , HLA-G Antigens/genetics , HLA-G Antigens/immunology , Humans , Ligands , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Isoforms , Receptors, Immunologic/metabolism , Structure-Activity Relationship , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
7.
J Neurosci Res ; 95(10): 2051-2058, 2017 10.
Article in English | MEDLINE | ID: mdl-28244141

ABSTRACT

The claudin family comprises four-pass transmembrane proteins involved in the formation of tight junctions (TJs). Relatively recently, claudin domain containing (CLDND) 1, also known as claudin-25, was identified as a novel member of the claudin family. In the present study, we revealed that in the adult murine brain, CLDND1 is abundant in the cerebellum among common sites of intracerebral hemorrhage. Thus, the dynamics of CLDND1 after cerebellar hemorrhage were examined. Both CLDND1 mRNA and protein levels transiently decreased at 24 hr after hemorrhagic insult. For immunostaining, an anti-CLDND1 antibody that recognizes the specific epitope in the extracellular first loop was prepared. Dual immunohistochemical staining with CD31 using coronal cryosections of intact murine cerebellum tissue revealed that CLDND1 is expressed on endothelial cells. We therefore performed an in vitro permeability test using a human brain endothelial cell (HBEC) line to reveal whether CLDND1 contributes to cell adhesion like other claudins. CLDND1 was expressed on HBECs as well as in murine cerebellum tissue, and a strong signal was observed at TJs. RNA interference against CLDND1 decreased both the mRNA and protein levels without cytotoxicity. The permeability to small molecules, but not to large ones, across confluent HBECs increased on CLDND1 knockdown compared with mock-treated cells. These results suggest that the transient decrease of CLDND1 after cerebellar hemorrhage is responsible for low-molecular-weight selective vascular hyperpermeability. © 2017 Wiley Periodicals, Inc.


Subject(s)
Cell Adhesion/physiology , Cerebral Hemorrhage/pathology , Claudins/metabolism , Endothelial Cells/metabolism , Animals , Capillary Permeability/physiology , Cells, Cultured , Cerebellum/metabolism , Cerebellum/pathology , Endothelial Cells/pathology , Humans , Male , Mice
8.
J Food Sci ; 82(5): 1224-1230, 2017 May.
Article in English | MEDLINE | ID: mdl-28346686

ABSTRACT

The ability of catechins and their related compounds to inhibit breast cancer resistance protein (BCRP) function in Caco-2 cell monolayers was investigated with mitoxantrone as a BCRP substrate. The gallate or pyrogallol moiety on the catechin structure seemed to promote increased cellular accumulation and inhibit efflux transport of mitoxantrone. The ability of gallate catechins such as (-)-epigallocatechin gallate (EGCG) and (-)-epicatechin gallate (ECG) to increase cellular accumulation and inhibit efflux transport of mitoxantrone was greater than that of nongallate catechins. Gallic acid octyl ester (GAO) also increased intracellular mitoxantrone accumulation. Experiments using GAO derivatives indicated that the gallate moiety required the presence of a long carbon chain for BCRP inhibition. Cellular accumulation and reduced efflux transport of mitoxantrone were greater with epigallocatechin 3-(3″-O-butyl) gallate than with EGCG. EGCG inhibition of BCRP seemed to be restricted by hydrophobicity. The co-administration of catechins, particularly EGCG and related compounds, with greater hydrophobicity may increase the therapeutic activities of BCRP substrates such as mitoxantrone.


Subject(s)
Analgesics/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Mitoxantrone/metabolism , Biological Transport/drug effects , Caco-2 Cells , Humans , Tea/chemistry
9.
Biochem Biophys Res Commun ; 486(2): 476-480, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28315682

ABSTRACT

Both cholesterol and α-tocopherol are essential lipophilic nutrients for humans and animals. Although cholesterol in excess causes severe problems such as coronary heart disease, it is a necessary component of cell membranes and is the precursor for the biosynthesis of steroid hormones and bile acids. Niemann-Pick C1-like 1 (NPC1L1) is a cholesterol transporter that is highly expressed in the small intestine and liver in humans and plays an important role in cholesterol homeostasis. Cholesterol promotes NPC1L1 endocytosis, which is an early step in cholesterol uptake. Furthermore, α-tocopherol is the most active form of vitamin E, and sufficient amounts of vitamin E are critical for health. It has been reported that NPC1L1 mediates α-tocopherol absorption; however, the mechanisms underlying this process are unknown. In this study, we found that treatment of cells that stably express NPC1L1-GFP with α-tocopherol promotes NPC1L1 endocytosis, and the NPC1L1 inhibitor, ezetimibe, efficiently prevents the α-tocopherol-induced endocytosis of NPC1L1. Cholesterol binding to the N-terminal domain (NTD) of NPC1L1 (NPC1L1-NTD) is essential for NPC1L1-mediated cholesterol absorption. We found that α-tocopherol competitively binds NPC1L1-NTD with cholesterol. Furthermore, when cells stably expressed NPC1L1ΔNTD-GFP, α-tocopherol could not induce the endocytosis of NPC1L1ΔNTD. Taken together, these results demonstrate that NPC1L1 recognizes α-tocopherol via its NTD and mediates α-tocopherol uptake through the same mechanism as cholesterol absorption.


Subject(s)
Cholesterol/metabolism , Hepatocytes/metabolism , Membrane Proteins/metabolism , Recombinant Fusion Proteins/metabolism , alpha-Tocopherol/metabolism , Animals , Anticholesteremic Agents/pharmacology , Binding, Competitive , Biological Transport , Caco-2 Cells , Cell Line, Tumor , Cholesterol/pharmacology , Endocytosis/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Ezetimibe/pharmacology , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Transport Proteins , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Domains , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
10.
Pharmacol Rep ; 68(3): 649-53, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27031052

ABSTRACT

BACKGROUND: Prazosin is an α1 adrenoceptor antagonist used in pharmacotherapy for the treatment of hypertension. Prazosin alters lipid metabolism in vivo, but the involved mechanism is not fully understood. In this study, we investigated the mechanism underlying the alteration of lipid metabolism. We show that the prazosin-stimulated release of hepatic triacylglyceride lipase (HTGL) from primary cultured rat hepatocytes involved Ca(2+)/calmodulin-dependent protein kinase II (CaMK-II) activation. METHODS: Primary cultured rat hepatocytes were incubated with prazosin and other agents. The hepatocytes were used in the CaMK-II and protein kinase A (PKA) activity assay. The supernatant was used in the HTGL activity assay and western blotting. RESULTS: Prazosin-stimulated HTGL release was suppressed by the inositol triphosphate receptor inhibitor xestospongin C and by the calmodulin inhibitor trifluoperazine but not by the protein kinase C inhibitor chelerythrine chloride or a diacylglycerol kinase inhibitor (R59949). Furthermore, the calmodulin-dependent protein kinase II (CaMK-II) activity in prazosin-treated hepatocytes increased in a time- and dose-dependent manner. The cAMP-dependent PKA activity of prazosin-stimulated hepatocytes was suppressed by a phospholipase C (PLC) inhibitor (U-73122), trifluoperazine, and a CaMK-II inhibitor (KN-93). CONCLUSIONS: These results suggested that prazosin-stimulated HTGL release from hepatocytes was caused by activation of PKA associated with stimulation of CaMK-II activity through a signal cascade from PLC.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Hepatocytes/metabolism , Lipoprotein Lipase/metabolism , Prazosin/pharmacology , Animals , Benzophenanthridines/pharmacology , Benzylamines/pharmacology , Dose-Response Relationship, Drug , Estrenes/pharmacology , Macrocyclic Compounds/pharmacology , Male , Oxazoles/pharmacology , Piperidines/pharmacology , Prazosin/antagonists & inhibitors , Primary Cell Culture , Pyrrolidinones/pharmacology , Quinazolinones/pharmacology , Rats , Sulfonamides/pharmacology , Time Factors , Trifluoperazine/pharmacology
11.
Eur J Immunol ; 45(6): 1605-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25826155

ABSTRACT

Emerging evidence has revealed the pivotal roles of C-type lectin-like receptors (CTLRs) in the regulation of a wide range of immune responses. Human natural killer cell receptor-P1A (NKRP1A) is one of the CTLRs and recognizes another CTLR, lectin-like transcript 1 (LLT1) on target cells to control NK, NKT and Th17 cells. The structural basis for the NKRP1A-LLT1 interaction was limitedly understood. Here, we report the crystal structure of the ectodomain of LLT1. The plausible receptor-binding face of the C-type lectin-like domain is flat, and forms an extended ß-sheet. The residues of this face are relatively conserved with another CTLR, keratinocyte-associated C-type lectin, which binds to the CTLR member, NKp65. A LLT1-NKRP1A complex model, prepared using the crystal structures of LLT1 and the keratinocyte-associated C-type lectin-NKp65 complex, reasonably satisfies the charge consistency and the conformational complementarity to explain a previous mutagenesis study. Furthermore, crystal packing and analytical ultracentrifugation revealed dimer formation, which supports a complex model. Our results provide structural insights for understanding the binding modes and signal transduction mechanisms, which are likely to be conserved in the CTLR family, and for further rational drug design towards regulating the LLT1 function.


Subject(s)
Lectins, C-Type/chemistry , Models, Molecular , NK Cell Lectin-Like Receptor Subfamily B/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Cell Surface/chemistry , Amino Acid Sequence , Binding Sites , Crystallization , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Molecular Sequence Data , Mutation , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Protein Binding , Protein Multimerization , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Alignment
12.
Proc Natl Acad Sci U S A ; 110(43): 17438-43, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24101491

ABSTRACT

Mincle [macrophage inducible Ca(2+)-dependent (C-type) lectin; CLEC4E] and MCL (macrophage C-type lectin; CLEC4D) are receptors for the cord factor TDM (trehalose-6,6'-dimycolate), a unique glycolipid of mycobacterial cell-surface components, and activate immune cells to confer adjuvant activity. Although it is known that receptor-TDM interactions require both sugar and lipid moieties of TDM, the mechanisms of glycolipid recognition by Mincle and MCL remain unclear. We here report the crystal structures of Mincle, MCL, and the Mincle-citric acid complex. The structures revealed that these receptors are capable of interacting with sugar in a Ca(2+)-dependent manner, as observed in other C-type lectins. However, Mincle and MCL uniquely possess shallow hydrophobic regions found adjacent to their putative sugar binding sites, which reasonably locate for recognition of fatty acid moieties of glycolipids. Functional studies using mutant receptors as well as glycolipid ligands support this deduced binding mode. These results give insight into the molecular mechanism of glycolipid recognition through C-type lectin receptors, which may provide clues to rational design for effective adjuvants.


Subject(s)
Cord Factors/chemistry , Lectins, C-Type/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Immunologic/chemistry , Amino Acid Sequence , Animals , Binding Sites/genetics , Calcium/chemistry , Calcium/metabolism , Citric Acid/chemistry , Citric Acid/metabolism , Cord Factors/metabolism , Crystallography, X-Ray , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Mice , Molecular Sequence Data , Mutation , Protein Binding , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance
13.
J Pharm Pharmacol ; 65(7): 1037-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23738731

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the transporter-mediated transport of N-acetyl 5-aminosalicylic acid (Ac-5-ASA) and the effect of quercetin on Ac-5-ASA transport. METHODS: Caco-2 cell monolayers grown in Transwells were used to study the transport of Ac-5-ASA in the absence or presence of quercetin, and apical-to-basolateral and basolateral-to-apical apparent permeability (PappAB and PappBA values, respectively) was determined. The effect of transporter inhibitors, such as MK571, quinidine and mitoxantrone, on the transport of Ac-5-ASA was investigated. KEY FINDINGS: In the absence of transporter mediators, the transport of Ac-5-ASA was much higher in the basolateral-to-apical direction than in the opposite direction. The PappBA/PappAB ratio of Ac-5-ASA was 4.89. Quercetin inhibited the apical efflux of Ac-5-ASA and decreased the PappBA/PappAB ratio to 1.05. Of the transporter inhibitors, MK571 decreased the PappBA/PappAB ratio to 1.07; however, neither quinidine nor mitoxantrone had an effect on Ac-5-ASA transport. CONCLUSIONS: Ac-5-ASA was excreted by multidrug resistance-associated protein 2 from Caco-2 cells, and its transport was inhibited by quercetin. Our findings suggest that dose levels of sulfasalazine or 5-aminosalicylic acid can be decreased by coadministration of quercetin, leading to improved pharmaceutical care for inflammatory bowel diseases.


Subject(s)
Aminosalicylic Acids/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Quercetin/pharmacology , Biological Transport , Caco-2 Cells , Drug Interactions , Humans , Mitoxantrone/pharmacology , Multidrug Resistance-Associated Protein 2 , Propionates/pharmacology , Quinidine/pharmacology , Quinolines/pharmacology
14.
Protein Pept Lett ; 19(4): 468-73, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21933116

ABSTRACT

Measles virus (MV), one of the most contagious agents, infects immune cells using the signaling lymphocyte activation molecule (SLAM) on the cell surface. A complex of SLAM and the attachment protein, hemagglutinin (MVH), has remained elusive due to the intrinsic handling difficulty including glycosylation. Furthermore, crystals obtained of this complex are either nondiffracting or poorly-diffracting. To solve this problem, we designed a systematic approach using a combination of the following techniques; (1) a transient expression system in HEK293SGnTI(-) cells, (2) lysine methylation, (3) structure-guided mutagenesis directed at better crystal packing, (4) Endo H treatment, (5) single-chain formation for stable complex, and (6) floating-drop vapor diffusion. Using our approach, the receptor-binding head domain of MV-H covalently fused with SLAM was successfully crystallized and diffraction was improved from 4.5 Å to a final resolution of 3.15 Å . These combinational methods would be useful as crystallization strategies for complexes of glycoproteins and their receptors.


Subject(s)
Antigens, CD/chemistry , Crystallization/methods , Hemagglutinins/chemistry , Receptors, Cell Surface/chemistry , Glycoproteins/chemistry , HEK293 Cells , Humans , Macromolecular Substances/chemistry , Measles virus/chemistry , Methylation , Signaling Lymphocytic Activation Molecule Family Member 1
15.
J Biol Chem ; 286(27): 23823-30, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21572041

ABSTRACT

Human Th17 cells express high levels of CD161, a member of the killer cell lectin-like receptor (KLR) family (also referred to as NK receptor-P1A (NKRP1A) or KLRB1), as a representative marker. CD161 is also expressed on natural killer (NK) cells and NKT cells. Lectin-like transcript 1 (LLT1), another KLR family member, was recently identified as a ligand for CD161. This interaction may play pivotal roles in the immunomodulatory functions of Th17 cells as well as those of NK and NKT cells. However, the molecular basis for the interaction is poorly understood. Here we show that the extracellular domain of CD161 bound directly to LLT1 with a K(d) of 48 µM and with the fast kinetics typical of cell-cell recognition receptors. Mutagenesis revealed that the similar membrane-distal ß-sheet and loop regions of both CD161 and LLT1 were utilized for the binding, and notably, these regions correspond to the ligand-binding sites for major histocompatibility complex (MHC)-recognizing KLRs. Furthermore, we found a pair of detrimental mutations for both molecules that restored the binding. These results reveal a new template model for the recognition mode between the KLR family members and provide insights into the molecular mechanism underlying Th17/NK/NKT-mediated immune responses.


Subject(s)
Lectins, C-Type/metabolism , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Cell Surface/metabolism , HEK293 Cells , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mutation , NK Cell Lectin-Like Receptor Subfamily B/genetics , NK Cell Lectin-Like Receptor Subfamily B/immunology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
16.
Nat Struct Mol Biol ; 18(2): 135-41, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21217702

ABSTRACT

Measles virus, a major cause of childhood morbidity and mortality worldwide, predominantly infects immune cells using signaling lymphocyte activation molecule (SLAM) as a cellular receptor. Here we present crystal structures of measles virus hemagglutinin (MV-H), the receptor-binding glycoprotein, in complex with SLAM. The MV-H head domain binds to a ß-sheet of the membrane-distal ectodomain of SLAM using the side of its ß-propeller fold. This is distinct from attachment proteins of other paramyxoviruses that bind receptors using the top of their ß-propeller. The structure provides templates for antiviral drug design, an explanation for the effectiveness of the measles virus vaccine, and a model of the homophilic SLAM-SLAM interaction involved in immune modulations. Notably, the crystal structures obtained show two forms of the MV-H-SLAM tetrameric assembly (dimer of dimers), which may have implications for the mechanism of fusion triggering.


Subject(s)
Antigens, CD/metabolism , Hemagglutinins, Viral/chemistry , Hemagglutinins, Viral/metabolism , Measles virus/metabolism , Measles/virology , Receptors, Cell Surface/metabolism , Antigens, CD/chemistry , Cell Line , Crystallography, X-Ray , Humans , Measles virus/chemistry , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, Cell Surface/chemistry , Signaling Lymphocytic Activation Molecule Family Member 1
17.
J Biol Chem ; 285(7): 4941-50, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20007322

ABSTRACT

Oligosaccharyltransferase (OST) catalyzes the transfer of an oligosaccharide from a lipid donor to an asparagine residue in nascent polypeptide chains. In the bacterium Campylobacter jejuni, a single-subunit membrane protein, PglB, catalyzes N-glycosylation. We report the 2.8 A resolution crystal structure of the C-terminal globular domain of PglB and its comparison with the previously determined structure from the archaeon Pyrococcus AglB. The two distantly related oligosaccharyltransferases share unexpected structural similarity beyond that expected from the sequence comparison. The common architecture of the putative catalytic sites revealed a new catalytic motif in PglB. Site-directed mutagenesis analyses confirmed the contribution of this motif to the catalytic function. Bacterial PglB and archaeal AglB constitute a protein family of the catalytic subunit of OST along with STT3 from eukaryotes. A structure-aided multiple sequence alignment of the STT3/PglB/AglB protein family revealed three types of OST catalytic centers. This novel classification will provide a useful framework for understanding the enzymatic properties of the OST enzymes from Eukarya, Archaea, and Bacteria.


Subject(s)
Archaeal Proteins/chemistry , Bacterial Proteins/chemistry , Campylobacter jejuni/enzymology , Hexosyltransferases/chemistry , Membrane Proteins/chemistry , Pyrococcus/enzymology , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Hexosyltransferases/genetics , Hexosyltransferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology
18.
Curr Pharm Des ; 15(28): 3318-24, 2009.
Article in English | MEDLINE | ID: mdl-19860681

ABSTRACT

Human leukocyte antigen-G (HLA-G) is a non-classical HLA class I molecule, which was first discovered in 1987 by Geraghty and colleagues. While classical HLA class I molecules are expressed on all nucleated cells, the expression of the HLA-G molecule is highly tissue-restricted, such as to placental trophoblast cells. HLA-G binds inhibitory receptors such as leukocyte immunoglobulin-like receptors B1 (LILRB1/ILT2/CD85j) and LILRB2 (ILT4/CD85d), which are widely expressed on immune cells, to suppress a broad range of immune responses. Thus, the expression of HLA-G in placenta protects the fetus from the maternal immune system. On the other hand, emerging studies have shown the relevance of the HLA-G molecule in pathologic conditions, such as transplantation rejection, autoimmunity, and cancer. HLA-G has other unique characteristics, in contrast with classical HLA molecules, including the existence of various forms of HLA-G: several splice variants, subunit-deficient conformations, homodimers, and their combinations have been found. In this review, we highlight the molecular basis for the tolerogenic ability of the HLA-G molecule, especially by LILR recognition of various forms of HLA-G. We also discuss the potential clinical applications of HLA-G molecules.


Subject(s)
HLA Antigens/chemistry , Histocompatibility Antigens Class I/chemistry , Animals , Antigens, CD/chemistry , Antigens, CD/immunology , Female , Fetus/immunology , HLA Antigens/genetics , HLA Antigens/immunology , HLA-G Antigens , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Leukocyte Immunoglobulin-like Receptor B1 , Polymorphism, Genetic/genetics , Pregnancy , Receptors, Antigen/chemistry , Receptors, Antigen/immunology , Receptors, Immunologic/chemistry , Receptors, Immunologic/immunology , Receptors, KIR/chemistry , Receptors, KIR/immunology
19.
EMBO J ; 27(1): 234-43, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18046457

ABSTRACT

Asn-glycosylation is widespread not only in eukaryotes but also in archaea and some eubacteria. Oligosaccharyltransferase (OST) catalyzes the co-translational transfer of an oligosaccharide from a lipid donor to an asparagine residue in nascent polypeptide chains. Here, we report that a thermophilic archaeon, Pyrococcus furiosus OST is composed of the STT3 protein alone, and catalyzes the transfer of a heptasaccharide, containing one hexouronate and two pentose residues, onto peptides in an Asn-X-Thr/Ser-motif-dependent manner. We also determined the 2.7-A resolution crystal structure of the C-terminal soluble domain of Pyrococcus STT3. The structure-based multiple sequence alignment revealed a new motif, DxxK, which is adjacent to the well-conserved WWDYG motif in the tertiary structure. The mutagenesis of the DK motif residues in yeast STT3 revealed the essential role of the motif in the catalytic activity. The function of this motif may be related to the binding of the pyrophosphate group of lipid-linked oligosaccharide donors through a transiently bound cation. Our structure provides the first structural insights into the formation of the oligosaccharide-asparagine bond.


Subject(s)
Catalytic Domain/physiology , Hexosyltransferases/chemistry , Membrane Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Carbohydrate Sequence , Catalytic Domain/genetics , Crystallography, X-Ray , Hexosyltransferases/genetics , Hexosyltransferases/isolation & purification , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/genetics , Pyrococcus furiosus/enzymology , Pyrococcus furiosus/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics
20.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 63(Pt 9): 798-801, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17768359

ABSTRACT

Oligosaccharyltransferase catalyzes the transfer of preassembled oligosaccharides onto asparagine residues in nascent polypeptide chains. The STT3 subunit is thought to bear the catalytic site. The C-terminal domain of the STT3 protein of Pyrococcus furiosus was expressed in Escherichia coli cells. STT3 protein prepared from two different sources, the soluble fraction and the inclusion bodies, produced crystals that diffracted to 2.7 A. During crystallization screening, cocrystals of P. furiosus STT3 with an E. coli 50S ribosomal protein, L7/L12, were accidentally obtained. This cross-species interaction is not biologically relevant, but may be used to design a built-in polypeptide substrate for the STT3 crystals.


Subject(s)
Archaeal Proteins/chemistry , Hexosyltransferases/chemistry , Membrane Proteins/chemistry , Pyrococcus furiosus/enzymology , Archaeal Proteins/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Campylobacter/enzymology , Glycosylation , Hexosyltransferases/genetics , Hexosyltransferases/isolation & purification , Hexosyltransferases/metabolism , Hot Temperature , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Protein Folding , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...