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










Publication year range
1.
Methods Enzymol ; 675: 235-273, 2022.
Article in English | MEDLINE | ID: mdl-36220272

ABSTRACT

Protein phosphorylation is the most common post-translational modification of proteins and functions as a molecular switch for their regulation. This modification is reversibly regulated by protein kinases and phosphatases. In most cases, the phosphorylation of enzymes positively or negatively regulates enzyme activity. However, we found that the phosphorylation of DDHD1 phospholipase A1 (PLA1) did not affect PLA1 activity. Integrated analyses, including phospho-proteomics, Phos-tag SDS-PAGE, PLA1 enzyme assays, and immunofluorescent microscopy, revealed the subcellular localization of DDHD1 without greatly affecting its PLA1 activity. Our findings may contribute to understanding rare clinical cases that concern the implications of protein phosphorylation.


Subject(s)
Phosphoric Monoester Hydrolases , Protein Kinases , Humans , Phospholipases A1/genetics , Phosphorylation
2.
Int J Mol Sci ; 23(18)2022 Sep 18.
Article in English | MEDLINE | ID: mdl-36142844

ABSTRACT

We previously reported that lysophosphatidylinositol (LPI) functions as an endogenous agonist of GPR55, a novel cannabinoid receptor. However, the physiological roles of LPI-GPR55 have not yet been elucidated in detail. In the present study, we found that LPI induced morphological changes in GPR55-expressing HEK293 cells. LPI induced the cell rounding of GPR55-expressing HEK293 cells but not of empty-vector-transfected cells. LPI also induced the activation of small GTP-binding protein RhoA and increased stress fiber formation in GPR55-expressing HEK293 cells. The inhibition of RhoA and Rho kinase ROCK by the C3 exoenzyme and the ROCK inhibitor reduced LPI-induced cell rounding and stress fiber formation. These results clearly indicated that the LPI-induced morphological changes and the assembly of the cytoskeletons were mediated through the GPR55-RhoA-ROCK pathway.


Subject(s)
Receptors, G-Protein-Coupled , rho-Associated Kinases , HEK293 Cells , Humans , Lysophospholipids/metabolism , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/agonists , Stress Fibers/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
3.
J Virol ; 95(17): e0080721, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34106748

ABSTRACT

The membrane fusion between the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host cells is essential for the initial step of infection; therefore, the host cell membrane components, including sphingolipids, influence the viral infection. We assessed several inhibitors of the enzymes pertaining to sphingolipid metabolism, against SARS-CoV-2 spike protein (S)-mediated cell-cell fusion and viral infection. N-(4-Hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Δ4-desaturase 1 (DES1), suppressed cell-cell fusion and viral infection. The analysis of sphingolipid levels revealed that the inhibition efficiencies of cell-cell fusion and viral infection in 4-HPR-treated cells were consistent with an increased ratio of saturated sphinganine-based lipids to total sphingolipids. We investigated the relationship of DES1 with the inhibition efficiencies of cell-cell fusion. The changes in the sphingolipid profile induced by 4-HPR were mitigated by the supplementation with exogenous cell-permeative ceramide; however, the reduced cell-cell fusion could not be reversed. The efficiency of cell-cell fusion in DES1 knockout (KO) cells was at a level comparable to that in wild-type (WT) cells; however, the ratio of saturated sphinganine-based lipids to the total sphingolipids was higher in DES1 KO cells than in WT cells. 4-HPR reduced cell membrane fluidity without any significant effects on the expression or localization of angiotensin-converting enzyme 2, the SARS-CoV-2 receptor. Therefore, 4-HPR suppresses SARS-CoV-2 S-mediated membrane fusion through a DES1-independent mechanism, and this decrease in membrane fluidity induced by 4-HPR could be the major cause for the inhibition of SARS-CoV-2 infection. IMPORTANCE Sphingolipids could play an important role in SARS-CoV-2 S-mediated membrane fusion with host cells. We studied the cell-cell fusion using SARS-CoV-2 S-expressing cells and sphingolipid-manipulated target cells, with an inhibitor of the sphingolipid metabolism. 4-HPR (also known as fenretinide) is an inhibitor of DES1, and it exhibits antitumor activity and suppresses cell-cell fusion and viral infection. 4-HPR suppresses membrane fusion through a decrease in membrane fluidity, which could possibly be the cause for the inhibition of SARS-CoV-2 infection. There is accumulating clinical data on the safety of 4-HPR. Therefore, it could be a potential candidate drug against COVID-19.


Subject(s)
Cell Membrane/metabolism , Fenretinide/pharmacology , Membrane Fluidity/drug effects , Oxidoreductases/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Cell Fusion , Cell Membrane/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Membrane Fluidity/genetics , Oxidoreductases/deficiency , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics
4.
J Biol Chem ; 297(1): 100851, 2021 07.
Article in English | MEDLINE | ID: mdl-34089703

ABSTRACT

Phospholipase A1 (PLA1) hydrolyzes the fatty acids of glycerophospholipids, which are structural components of the cellular membrane. Genetic mutations in DDHD1, an intracellular PLA1, result in hereditary spastic paraplegia (HSP) in humans. However, the regulation of DDHD1 activity has not yet been elucidated in detail. In the present study, we examined the phosphorylation of DDHD1 and identified the responsible protein kinases. We performed MALDI-TOF MS/MS analysis and Phos-tag SDS-PAGE in alanine-substitution mutants in HEK293 cells and revealed multiple phosphorylation sites in human DDHD1, primarily Ser8, Ser11, Ser723, and Ser727. The treatment of cells with a protein phosphatase inhibitor induced the hyperphosphorylation of DDHD1, suggesting that multisite phosphorylation occurred not only at these major, but also at minor sites. Site-specific kinase-substrate prediction algorithms and in vitro kinase analyses indicated that cyclin-dependent kinase CDK1/cyclin A2 phosphorylated Ser8, Ser11, and Ser727 in DDHD1 with a preference for Ser11 and that CDK5/p35 also phosphorylated Ser11 and Ser727 with a preference for Ser11. In addition, casein kinase CK2α1 was found to phosphorylate Ser104, although this was not a major phosphorylation site in cultivated HEK293 cells. The evaluation of the effects of phosphorylation revealed that the phosphorylation mimic mutants S11/727E exhibit only 20% reduction in PLA1 activity. However, the phosphorylation mimics were mainly localized to focal adhesions, whereas the phosphorylation-resistant mutants S11/727A were not. This suggested that phosphorylation alters the subcellular localization of DDHD1 without greatly affecting its PLA1 activity.


Subject(s)
CDC2 Protein Kinase/genetics , Cyclin A2/genetics , Phospholipases A1/genetics , CDC2 Protein Kinase/chemistry , Cell Membrane/chemistry , Cell Membrane/genetics , Cyclin A2/chemistry , Glycerophospholipids/chemistry , Glycerophospholipids/genetics , HEK293 Cells , Humans , Phospholipases A1/chemistry , Phospholipases A1/metabolism , Phosphorylation/genetics , Spastic Paraplegia, Hereditary/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
J Biol Chem ; 293(45): 17505-17522, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30242129

ABSTRACT

Sphingolipids, including sphingomyelin (SM) and glucosylceramide (GlcCer), are generated by the addition of a polar head group to ceramide (Cer). Sphingomyelin synthase 1 (SMS1) and glucosylceramide synthase (GCS) are key enzymes that catalyze the conversion of Cer to SM and GlcCer, respectively. GlcCer synthesis has been postulated to occur mainly in cis-Golgi, and SM synthesis is thought to occur in medial/trans-Golgi; however, SMS1 and GCS are known to partially co-localize in cisternae, especially in medial/trans-Golgi. Here, we report that SMS1 and GCS can form a heteromeric complex, in which the N terminus of SMS1 and the C terminus of GCS are in close proximity. Deletion of the N-terminal sterile α-motif of SMS1 reduced the stability of the SMS1-GCS complex, resulting in a significant reduction in SM synthesis in vivo In contrast, chemical-induced heterodimerization augmented SMS1 activity, depending on an increase in the amount and stability of the complex. Fusion of the SMS1 N terminus to the GCS C terminus via linkers of different lengths increased SM synthesis and decreased GlcCer synthesis in vivo These results suggest that formation of the SMS1-GCS heteromeric complex increases SM synthesis and decreases GlcCer synthesis. Importantly, this regulation of relative Cer levels by the SMS1-GCS complex was confirmed by CRISPR/Cas9-mediated knockout of SMS1 or GCS combined with pharmacological inhibition of Cer transport protein in HEK293T cells. Our findings suggest that complex formation between SMS1 and GCS is part of a critical mechanism controlling the metabolic fate of Cer in the Golgi.


Subject(s)
Glucosylceramides/biosynthesis , Glucosyltransferases/metabolism , Membrane Proteins/metabolism , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/metabolism , Sphingomyelins/biosynthesis , Transferases (Other Substituted Phosphate Groups)/metabolism , trans-Golgi Network/enzymology , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Gene Knockdown Techniques , Glucosylceramides/genetics , Glucosyltransferases/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Multienzyme Complexes/genetics , Nerve Tissue Proteins/genetics , Sequence Deletion , Sphingomyelins/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , trans-Golgi Network/genetics
6.
Biology (Basel) ; 6(2)2017 Mar 30.
Article in English | MEDLINE | ID: mdl-28358327

ABSTRACT

The coenzyme A (CoA)-independent transacylation system catalyzes fatty acid transfer from phospholipids to lysophospholipids in the absence of cofactors such as CoA. It prefers to use C20 and C22 polyunsaturated fatty acids such as arachidonic acid, which are esterified in the glycerophospholipid at the sn-2 position. This system can also acylate alkyl ether-linked lysophospholipids, is involved in the enrichment of arachidonic acid in alkyl ether-linked glycerophospholipids, and is critical for the metabolism of eicosanoids and platelet-activating factor. Despite their importance, the enzymes responsible for these reactions have yet to be identified. In this review, we describe the features of the Ca2+-independent, membrane-bound CoA-independent transacylation system and its selectivity for arachidonic acid. We also speculate on the involvement of phospholipase A2 in the CoA-independent transacylation reaction.

7.
J Biol Chem ; 292(3): 1122-1141, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27927984

ABSTRACT

Sphingomyelin synthase (SMS) is the key enzyme for cross-talk between bioactive sphingolipids and glycerolipids. In mammals, SMS consists of two isoforms: SMS1 is localized in the Golgi apparatus, whereas SMS2 is localized in both the Golgi and plasma membranes. SMS2 seems to exert cellular functions through protein-protein interactions; however, the existence and functions of quaternary structures of SMS1 and SMS2 remain unclear. Here we demonstrate that both SMS1 and SMS2 form homodimers. The SMSs have six membrane-spanning domains, and the N and C termini of both proteins face the cytosolic side of the Golgi apparatus. Chemical cross-linking and bimolecular fluorescence complementation revealed that the N- and/or C-terminal tails of the SMSs were in close proximity to those of the other SMS in the homodimer. Homodimer formation was significantly decreased by C-terminal truncations, SMS1-ΔC22 and SMS2-ΔC30, indicating that the C-terminal tails of the SMSs are primarily responsible for homodimer formation. Moreover, immunoprecipitation using deletion mutants revealed that the C-terminal tail of SMS2 mainly interacted with the C-terminal tail of its homodimer partner, whereas the C-terminal tail of SMS1 mainly interacted with a site other than the C-terminal tail of its homodimer partner. Interestingly, homodimer formation occurred in the endoplasmic reticulum (ER) membrane before trafficking to the Golgi apparatus. Reduced homodimerization caused by C-terminal truncations of SMSs significantly reduced ER-to-Golgi transport. Our findings suggest that the C-terminal tails of SMSs are involved in homodimer formation, which is required for efficient transport from the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Multimerization/physiology , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , Golgi Apparatus/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Domains , Protein Transport/physiology , Transferases (Other Substituted Phosphate Groups)/genetics
8.
Biology (Basel) ; 3(4): 801-30, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25415055

ABSTRACT

Acyl-CoA:glycerol-3-phosphate acyltransferase (GPAT) and acyl-CoA: 1-acyl-glycerol-3-phosphate acyltransferase (AGPAT) are involved in the de novo synthesis of triacylglycerol (TAG) and glycerophospholipids. Many enzymes belonging to the GPAT/AGPAT family have recently been identified and their physiological or pathophysiological roles have been proposed. The roles of GPAT/AGPAT in the synthesis of TAG and obesity-related diseases were revealed through the identification of causative genes of these diseases or analyses of genetically manipulated animals. Recent studies have suggested that some isoforms of GPAT/AGPAT family enzymes are involved in the fatty acid remodeling of phospholipids. The enzymology of GPAT/AGPAT and their physiological/ pathological roles in the metabolism of glycerolipids have been described and discussed in this review.

9.
J Biol Chem ; 289(44): 30842-30856, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25231990

ABSTRACT

Membrane fusion between the viral envelope and plasma membranes of target cells has previously been correlated with HIV-1 infection. Lipids in the plasma membrane, including sphingomyelin, may be crucially involved in HIV-1 infection; however, the role of lipid-metabolic enzymes in membrane fusion remains unclear. In this study, we examined the roles of sphingomyelin synthase (SMS) in HIV-1 Env-mediated membrane fusion using a cell-cell fusion assay with HIV-1 mimetics and their target cells. We employed reconstituted cells as target cells that stably express Sms1 or Sms2 in Sms-deficient cells. Fusion susceptibility was ∼5-fold higher in Sms2-expressing cells (not in Sms1-expressing cells) than in Sms-deficient cells. The enhancement of fusion susceptibility observed in Sms2-expressing cells was reversed and reduced by Sms2 knockdown. We also found that catalytically nonactive Sms2 promoted membrane fusion susceptibility. Moreover, SMS2 co-localized and was constitutively associated with the HIV receptor·co-receptor complex in the plasma membrane. In addition, HIV-1 Env treatment resulted in a transient increase in nonreceptor tyrosine kinase (Pyk2) phosphorylation in Sms2-expressing and catalytically nonactive Sms2-expressing cells. We observed that F-actin polymerization in the region of membrane fusion was more prominent in Sms2-expressing cells than Sms-deficient cells. Taken together, our research provides insight into a novel function of SMS2 which is the regulation of HIV-1 Env-mediated membrane fusion via actin rearrangement.


Subject(s)
HIV-1/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Transferases (Other Substituted Phosphate Groups)/physiology , Virus Internalization , Actins/metabolism , Animals , Cell Membrane/enzymology , Cell Membrane/virology , Enzyme Activation , Focal Adhesion Kinase 2/metabolism , Gene Expression , Humans , Jurkat Cells , Mice, Knockout , Protein Multimerization , Protein Transport , Receptors, HIV/metabolism , Virus Attachment
10.
Prog Lipid Res ; 53: 18-81, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24125941

ABSTRACT

Over one hundred different phospholipid molecular species are known to be present in mammalian cells and tissues. Fatty acid remodeling systems for phospholipids including acyl-CoA:lysophospholipid acyltransferases, CoA-dependent and CoA-independent transacylation systems, are involved in the biosynthesis of these molecular species. Acyl-CoA:lysophospholipid acyltransferase system is involved in the synthesis of phospholipid molecular species containing sn-1 saturated and sn-2 unsaturated fatty acids. The CoA-dependent transacylation system catalyzes the transfer of fatty acids esterified in phospholipids to lysophospholipids in the presence of CoA without the generation of free fatty acids. The CoA-dependent transacylation reaction in the rat liver exhibits strict fatty acid specificity, i.e., three types of fatty acids (20:4, 18:2 and 18:0) are transferred. On the other hand, CoA-independent transacylase catalyzes the transfer of C20 and C22 polyunsaturated fatty acids from diacyl phospholipids to various lysophospholipids, especially ether-containing lysophospholipids, in the absence of any cofactors. CoA-independent transacylase is assumed to be involved in the accumulation of PUFA in ether-containing phospholipids. These enzymes are involved in not only the remodeling of fatty acids, but also the synthesis and degradation of some bioactive lipids and their precursors. In this review, recent progresses in acyltransferase research including the identification of the enzyme's genes are described.


Subject(s)
Acyltransferases/metabolism , Fatty Acids/chemistry , Phospholipids/chemistry , Phospholipids/metabolism , Acylation , Animals , Humans , Mammals , Phospholipids/biosynthesis
11.
J Biochem ; 154(5): 455-64, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23935187

ABSTRACT

LEC-1 is a major galectin in Caenorhabditis elegans and contains two carbohydrate recognition domains (CRDs), N-CRD and C-CRD. To determine the role of LEC-1, we examined the phenotypes of a mutant C. elegans strain lacking lec-1. We observed negligible differences in embryogenesis, morphogenesis and egg laying at 20 °C between the mutant and the wild-type. Furthermore, the life spans of the mutant and the wild-type were equivalent at either 20 °C or 25 °C. However, the lec-1 mutant showed a greater susceptibility to H2O2 and paraquat than the wild-type. This result suggests an increased susceptibility to oxidative stress, with the phenotypes being similar to those of lec-10 deletion mutants as previously described. To understand the molecular mechanism underlying this phenotype, C. elegans proteins bound by either the LEC-1 N-CRD or C-CRD were isolated and identified using a nano liquid chromatography-tandem mass spectrometry technique. MIG-6 was identified as a major binding partner of LEC-1 with both N- and C-CRD. From these results and previous reports, we speculate that interaction of LEC-1 and MIG-6 in the pharynx may affect susceptibility to paraquat and that LEC-10 has different functions from LEC-1 in regulating H2O2 and paraquat resistance in the intestine.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Galectins/metabolism , Oxidative Stress , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Chromatography, Liquid , Galectins/chemistry , Galectins/genetics , Hydrogen Peroxide/chemistry , Mutation , Nanotechnology , Phenotype , Tandem Mass Spectrometry
12.
Prostaglandins Other Lipid Mediat ; 107: 103-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23714700

ABSTRACT

Lysophosphatidylinositol (LPI) is a subspecies of lysophospholipid and is assumed to be not only a degradation product of phosphatidylinositol (PI), but also a bioactive lysophospholipid mediator. However, not much attention has been directed toward LPI compared to lysophosphatidic acid (LPA), since the receptor for LPI has not been identified. During screening for an agonist for the orphan G protein coupled receptor GPR55, we identified LPI, 2-arachidonoyl LPI in particular, as an agonist for GPR55. Our efforts to identify an LPI receptor facilitated research on LPI as a lipid messenger. In addition, we also found that DDHD1, previously identified as phosphatidic acid-preferring phospholipase A1, was one of the synthesizing enzymes of 2-arachidonoyl LPI. Here, we summarized the background for discovering the LPI receptor, and the actions/metabolism of LPI. We also referred to the biosynthesis of PI, a 1-stearoyl-2-arachidonoyl species, since the molecule is the precursor of 2-arachidonoyl LPI. Furthermore, we discussed physiological and/or pathophysiological processes involving LPI and GPR55, including the relevance of LPI-GPR55 and cannabinoids, since GPR55 was previously postulated to be another cannabinoid receptor. Although there is no doubt that GPR55 is the LPI receptor, we should re-consider whether or not GPR55 is in fact another cannabinoid receptor.


Subject(s)
Lysophospholipids/physiology , Receptors, G-Protein-Coupled/metabolism , Acetyltransferases/metabolism , Animals , Biosynthetic Pathways , Endocannabinoids/physiology , Humans , Lipid Metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/agonists , Signal Transduction
13.
Biol Pharm Bull ; 34(10): 1635-9, 2011.
Article in English | MEDLINE | ID: mdl-21963509

ABSTRACT

Galectins comprise a large family of ß-galactoside-binding proteins in animals and fungi. We previously isolated cDNAs of 10 galectin and galectin-like genes (lec-1 to lec-6 and lec-8 to lec-11) from Caenorhabditis elegans and characterized the carbohydrate-binding properties of their recombinant proteins. In the present study, we isolated cDNA corresponding to an open reading frame of the DC2.3a gene from C. elegans total RNA; this cDNA encodes another potential galectin. A recombinant DC2.3a protein was expressed in Escherichia coli and used for analysis. The protein displayed hemagglutinating activity against rabbit erythrocytes, bound to an asialofetuin-Sepharose column, and was eluted with lactose. Furthermore, frontal affinity chromatography (FAC) analysis confirmed that DC2.3a recognized oligosaccharides with a non-reducing terminal galactose. According to these results, we designated DC2.3 as lec-12. The carbohydrate-binding property of the recombinant DC2.3a/LEC-12a was essentially similar to that of LEC-6. Additionally, DC2.3a/LEC-12a and LEC-6 showed higher affinities for the galactoseß1→4fucose (Galß1→4Fuc) disaccharide than for N-acetyllactosamine. This suggests that the principal recognition unit is the Galß1→4Fuc disaccharide as in the case of the C. elegans galectins. However, the recombinant DC2.3a/LEC-12a showed weak affinity for N-glycan E3, which was previously shown to be a preferential endogenous ligand for LEC-6. The DC2.3a/LEC-12a endogenous ligand structures appear to be somewhat different but contain the same galactose-fucose recognition motif.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/chemistry , Disaccharides/genetics , Galactosides/genetics , Galectins/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Disaccharides/chemistry , Disaccharides/metabolism , Drug Evaluation, Preclinical , Escherichia coli/genetics , Escherichia coli/metabolism , Fluorescent Dyes/metabolism , Galactosides/chemistry , Galactosides/metabolism , Galectins/chemistry , Galectins/metabolism , Ligands , Molecular Targeted Therapy , Plasmids , Protein Binding , Rabbits
14.
Biol Pharm Bull ; 34(7): 1139-42, 2011.
Article in English | MEDLINE | ID: mdl-21720028

ABSTRACT

To study the endogenous counterpart of LEC-6, a major galectin in Caenorhabditis elegans, the proteomic analysis of glycoproteins captured by an immobilized LEC-6 column was performed using the nano liquid chromatography-tandem mass spectrometry (LC-MS/MS) technique. A protein recovered in a significant amount was determined to be either F57F4.3 or F57F4.4, although the method used could not determine which protein was the actual counterpart. Because the knockdown of the F57F4.3/4 genes in C. elegans is reported to cause growth retardation, we performed a double knockdown of the lec-6 and F57F4.3/4 genes. Although the RNA-mediated interference (RNAi) of lec-6 led to no obvious phenotype, the RNAi of both the lec-6 and F57F4.3/4 genes led to a significant reduction in growth rate when compared to the RNAi of F57F4.3/4 alone. Furthermore, to clarify which protein, F57F4.3 or F57F4.4, was responsible for the retarded growth, the levels of the F57F4.3/4 proteins expressed in a C. elegans wild type and a mutant lacking part of the F57F4.3 gene were compared. The levels of protein expressed by the wild type and the mutant were not significantly different, suggesting that the F57F4.3 protein contributes very little to growth retardation and that the major glycoprotein that interacts with LEC-6 is the F57F4.4 protein. These results suggest that binding with LEC-6 supports the function of F57F4.4 and that their cooperative functioning regulates the growth of C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Galectins/metabolism , Glycoproteins/metabolism , Animals , Base Sequence , Blotting, Western , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , DNA Primers , Galectins/genetics , Glycoproteins/genetics , Membrane Proteins , Polymerase Chain Reaction , Protein Binding , RNA Interference
15.
Glycoconj J ; 27(2): 267-76, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20077002

ABSTRACT

O-glycosylation of mucin is initiated by the attachment of N-acetyl-D-galactosamine (GalNAc) to serine or threonine residues in mucin core polypeptides by UDPGalNAc:polypeptide N-acetylgalactosaminyltransferases (ppGalNAc-Ts). It is not well understood how GalNAc attachment is regulated by multiple ppGalNAc-Ts in each cell. In the present study, the expression levels of murine ppGalNAc-Ts (mGalNAc-Ts), T1, T2, T3, T4, T6, and T7 were compared between mouse colon carcinoma colon 38 cells and variant SL4 cells, selected for their metastatic potentials, by using the competitive RT-PCR method. The expression levels of mGalNAc-T1, T2, and T7 were slightly higher in the SL4 cells than in the colon 38 cells, whereas the expression level of mGalNAc-T3 in the SL4 cells was 1.5% of that in the colon 38 cells. Products of enzymatic incorporations of GalNAc residues into FITCPTTTPITTTTK peptide by the use of microsome fractions of these cells as the enzyme source were separated and characterized for the number of attached GalNAc residues and their positions. The maximum number of attached GalNAc residues was 6 and 4 when the microsome fractions of the colon 38 cells and SL4 cells were used, respectively. When the microsome fractions of the colon 38 cells were treated with a polyclonal antibody raised against mGalNAc-T3, the maximum number of incorporated GalNAc residues was 4. These results strongly suggest that mGalNAc-T3 in colon 38 cells is involved in additional transfer of GalNAc residues to this peptide.


Subject(s)
Colonic Neoplasms/enzymology , Liver Neoplasms/secondary , N-Acetylgalactosaminyltransferases/metabolism , Acetylgalactosamine/metabolism , Amino Acid Sequence , Animals , Antibodies/pharmacology , Cell Line, Tumor , Colonic Neoplasms/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycopeptides/chemistry , Glycopeptides/metabolism , Glycosylation/drug effects , Humans , Lectins/metabolism , Mice , Microsomes/drug effects , Microsomes/enzymology , Molecular Sequence Data , Mucins/metabolism , N-Acetylgalactosaminyltransferases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity/drug effects , Polypeptide N-acetylgalactosaminyltransferase
16.
Biol Pharm Bull ; 32(12): 1973-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952414

ABSTRACT

Galectins are a family of beta-galactoside-binding lectins. They are involved in the regulation of a variety of biological phenomena in mammals. However, little is known about their roles in invertebrates. Caenorhabditis elegans is a well-characterized model organism whose complete genome has been sequenced. C. elegans is now being studied extensively in various fields of medical sciences. In this study, we examined the phenotypes of a mutant strain of C. elegans (tm1262) lacking lec-10, a galectin-encoding gene. We observed no difference in the rates of embryonic lethality and larval arrest/slow growth between this mutant strain and the wild-type strain. No apparent morphological defect was observed in the lec-10-deletion mutant (tm1262). Moreover, the life-spans of this mutant and the wild-type strain were equivalent. However, this mutant showed significantly greater susceptibility to paraquat and hydrogen peroxide than the wild type did. The lec-10-deletion mutants (tm1262) were as susceptible as the daf-16-deletion mutants (mu86) to paraquat and hydrogen peroxide. These results suggest that the deletion of lec-10 does not have a notable effect on the worm's survival under laboratory conditions. However, this study indicates that lec-10 does confer some protection against oxidative stress.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Gene Deletion , Oxidative Stress/genetics , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Galectins/genetics , Galectins/metabolism , Hydrogen Peroxide , Oxidants , Paraquat , Phenotype
17.
Biochim Biophys Acta ; 1780(10): 1131-42, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18675319

ABSTRACT

Galectins form a large family of beta-galactoside-binding proteins in metazoa and fungi. This report presents a comparative study of the functions of potential galectin genes found in the genome database of Caenorhabditis elegans. We isolated full-length cDNAs of eight potential galectin genes (lec-2-5 and 8-11) from a lambdaZAP cDNA library. Among them, lec-2-5 were found to encode 31-35-kDa polypeptides containing two carbohydrate-recognition domains similar to the previously characterized lec-1, whereas lec-8-11 were found to encode 16-27-kDa polypeptides containing a single carbohydrate-recognition domain and a C-terminal tail of unknown function. Recombinant proteins corresponding to lec-1-4, -6, and 8-10 were expressed in Escherichia coli, and their sugar-binding properties were assessed. Analysis using affinity adsorbents with various beta-galactosides, i.e., N-acetyllactosamine (Galbeta1-4GlcNAc), lacto-N-neotetraose (Galbeta1-4GlcNAcbeta1-3Galbeta1-4Glc), and asialofetuin, demonstrated that LEC-1-4, -6, and -10 have a significant affinity for beta-galactosides, while the others have a relatively lower affinity. These results indicate that the integrity of key amino acid residues responsible for recognition of lactose (Galbeta1-4Glc) or N-acetyllactosamine in vertebrate galectins is also required in C. elegans galectins. However, analysis of their fine oligosaccharide-binding properties by frontal affinity chromatography suggests their divergence towards more specialized functions.


Subject(s)
Caenorhabditis elegans/metabolism , Carbohydrate Metabolism , Galectins/chemistry , Galectins/metabolism , Alternative Splicing , Amino Acid Sequence , Amino Acid Substitution , Animals , Caenorhabditis elegans/genetics , Chromatography, Affinity , Cloning, Molecular , Conserved Sequence , DNA, Complementary , Galectins/genetics , Galectins/isolation & purification , Genes, Helminth , Kinetics , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Phylogeny , Protein Sorting Signals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment
18.
Int J Cancer ; 112(6): 927-33, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15386364

ABSTRACT

In order to clarify the roles of tumor necrosis factor (TNF)-alpha in lung metastasis, we injected Renca cells intravenously into TNF receptor p55-deficient (TNF-Rp55 KO) and wild-type (WT) mice. Microscopic and macroscopic metastasis foci appeared in lungs at 7 and 14 days after the tumor injection, respectively. Moreover, metastasis foci expanded at similar rates in both WT and TNF-Rp55 KO mice until 21 days, and lungs were occupied with metastasis foci. However, later than 21 days after the injection, metastasis foci spontaneously regressed in TNF-Rp55 KO mice, whereas WT mice exhibited a progressive growth of metastasis foci. Moreover, metastasis foci remained reduced sizes in TNF-Rp55 KO mice even at 26 days, when all WT mice died with lungs filled with metastasis foci. Later than 21 days after the tumor injection, the number of apoptotic tumor cells was increased in TNF-Rp55 KO mice. In contrast, neovascularization was less evident in TNF-Rp55 KO than WT mice, with depressed hepatocyte growth factor (HGF) gene in TNF-Rp55 KO mice at 21 days after the tumor injection. Thus, TNF-Rp55-mediated signals can maintain tumor neovascularization at least partly by inducing HGF expression, and eventually support lung metastasis process.


Subject(s)
Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/secondary , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Neovascularization, Pathologic/prevention & control , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Carcinoma, Renal Cell/blood supply , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Injections, Intravenous , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lung Neoplasms/blood supply , Mice , Mice, Inbred BALB C , Mice, Knockout , Neovascularization, Pathologic/drug therapy , Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
19.
Int Immunopharmacol ; 3(5): 627-42, 2003 May.
Article in English | MEDLINE | ID: mdl-12757733

ABSTRACT

We previously observed that IL-4 gene transduction into a mouse colon 26 adenocarcinoma cell line abrogated its tumorigenicity due to the generation of anti-tumor CTL. DEC-205- and CD11c-double positive cells were increased in the lymph nodes of mice injected with IL-4-transfected cells between 2 and 3 days after the tumor injection, compared with those injected with parental cells. Most of these double positive cells expressed CD86 antigen. Among the chemokines with chemotactic activities against dendritic cells, monocyte chemoattractant protein (MCP)-1/CCL2, ABCD-1/CCL22, and liver and activation-regulated chemokine (LARC)/CCL20 gene expression was enhanced no later than 3 days after the tumor injection, in the draining lymph nodes of IL-4-transfected cell bearing mice. Moreover, gene expression of the receptor for MCP-1/CCL2, CCR2, was enhanced in the draining lymph nodes of the mice injected with IL-4-transfected cells, and most DEC-205-positive cells in the lymph nodes expressed CCR2. Finally, the administration of anti-MCP-1/CCL2 antibodies retarded the rate of tumor regression in mice injected with IL-4-tranfected cells, concomitantly with a decrease in DEC-205- and CD11c-double positive cell number in the draining lymph nodes. Thus, locally produced MCP-1/CCL2 may be responsible for IL-4-mediated tumor rejection presumably based on the induction of dendritic cell migration into the draining lymph nodes.


Subject(s)
Adenocarcinoma/immunology , Chemokine CCL2/physiology , Colonic Neoplasms/immunology , Dendritic Cells/physiology , Interleukin-4/physiology , Lymph Nodes/cytology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies/chemistry , Antibodies/immunology , Cell Line, Tumor , Cell Movement , Chemokines/biosynthesis , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor , Immunohistochemistry , Interleukin-4/genetics , Mice , Mice, Inbred BALB C , Receptors, Chemokine/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
Am J Pathol ; 162(4): 1249-58, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12651617

ABSTRACT

Hepatoma cell lines can produce a massive amount of chemokines in response to various stimuli including hepatitis viruses and their products. However, it remains elusive on the types of chemokine receptor(s) expressed in the hepatoma tissues and its roles in hepatoma development. To clarify these points, we examined the chemokine receptor expression in six human hepatoma cell lines. All of the hepatoma cell lines constitutively and exclusively expressed CCR1 mRNA and its protein on their cell surface. CCR1 expression was also detected on hepatoma cells and to a lesser degree, on endothelial cells in hepatoma tissues but not in normal liver tissues. Furthermore, CCL3 expression was detected in hepatoma cells, endothelial cells, and to a lesser degree, fibroblast-like cells in hepatoma tissue, whereas only occasional vascular endothelial cells and inflammatory cells in normal liver tissues were weakly positive for CCL3. Moreover, the forskolin-mediated increases in intracellular cAMP concentrations were inhibited by the ligands for CCR1, CCL3, CCL4, and CCL5, suggesting that the expressed CCR1 was functional. Four hepatoma cell lines produced CCL3 only in response to interleukin (IL)-1 alpha and IL-1 beta. Finally, IL-1 alpha and IL-1 beta were detected abundantly in hepatoma tissues but not in normal liver tissues. Thus, IL-1 may enhance the local production of CCL3, which may interact with CCR1 expressed on hepatoma cells, in an autocrine and/or paracrine manner.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Interleukin-1/physiology , Liver Neoplasms/metabolism , Macrophage Inflammatory Proteins/metabolism , Receptors, Chemokine/metabolism , Base Sequence , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Chemokine CCL3 , Chemokine CCL4 , DNA Primers , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Macrophage Inflammatory Proteins/genetics , Polymerase Chain Reaction , RNA, Messenger/genetics , Receptors, CCR1 , Receptors, Chemokine/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...