Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Alzheimers Dis ; 74(3): 741-746, 2020.
Article in English | MEDLINE | ID: mdl-32083579

ABSTRACT

Accumulation of advanced glycation end products (AGEs) has been linked with cognitive decline as a risk factor based on the analysis in small populations. We investigated the association between skin autofluorescence of AGEs and global cognitive function in a Japanese older (≥60 years) population (n = 4,041). The AGEs quartiles were inversely associated with the Revised Hasegawa's Dementia Scale score (Q1: reference, Q2: ß= -0.011, p = 0.537, Q3: ß= -0.043, p = 0.016, Q4: ß= -0.064, p < 0.001) independent of major risk factors. Accumulation of AGEs was associated with lower cognitive performance in older adults.


Subject(s)
Cognition , Glycation End Products, Advanced/metabolism , Psychomotor Performance , Aged , Cognition Disorders/epidemiology , Cognition Disorders/metabolism , Cognition Disorders/psychology , Cross-Sectional Studies , Educational Status , Female , Fingers/anatomy & histology , Fluorescence , Humans , Japan/epidemiology , Male , Middle Aged , Neuropsychological Tests , Risk Factors , Skin
2.
J Gerontol A Biol Sci Med Sci ; 74(9): 1446-1453, 2019 08 16.
Article in English | MEDLINE | ID: mdl-30329028

ABSTRACT

BACKGROUND: The accumulation of advanced glycation end product (AGE) might exert deleterious effects on musculoskeletal properties. Our study aims to clarify this possible association in a large general population. METHODS: This study investigated a general population of 9,203 patients (mean age, 57.8 years). Skeletal muscle mass was measured by bioelectrical impedance analysis, whereas accumulation of AGEs was assessed by skin autofluorescence (SAF-AGE). The muscle strength of upper and lower limbs and usual gait speed were measured in a portion of older (≥60 years of age) participants (n = 1,934). The speed of sound (SOS) in the calcaneal bone was assessed via a quantitative ultrasound technique. RESULTS: In the total population, the frequency of low skeletal muscle mass linearly increased with the SAF-AGE quartiles (Q1: 14.2%, Q2: 16.1%, Q3: 21.1%, Q4: 24.8%; p < .001), and this association was independent of covariates including glycemic traits (Q4: odds ratio [OR] = 1.48, p < .001). The association between the highest SAF-AGE quartile and low skeletal muscle mass remained significant in the older subpopulation (OR = 1.85, p = .002). A similar but weak association was observed for low SOS (Q1: 8.9%, Q2: 8.3%, Q3: 10.4%, Q4: 12.2%; p < .001). Similar inverse associations were also observed with grip strength (OR = 1.98, p = .003), hip flexion strength (OR = 1.50, p = .012), and hip abduction strength (OR = 1.78, p = .001), but not with usual gait speed. CONCLUSION: Accumulation of AGEs might be a deleterious factor for musculoskeletal properties.


Subject(s)
Bone Density , Geriatric Assessment , Glycation End Products, Advanced/metabolism , Muscle Strength , Muscle, Skeletal/anatomy & histology , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Organ Size
3.
Methods ; 154: 10-20, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30326272

ABSTRACT

The antibody drug market is rapidly expanding, and various antibody engineering technologies are being developed to create antibodies that can provide better benefit to patients. Although bispecific antibody drugs have been researched for more than 30 years, currently only a limited number of bispecific antibodies have achieved regulatory approval. Of the few successful examples of industrially manufacturing a bispecific antibody, the "common light chain format" is an elegant technology that simplifies the purification of a whole IgG-type bispecific antibody. Using this IgG format, the bispecific function can be introduced while maintaining the natural molecular shape of the antibody. In this article, we will first introduce the outline, prospects, and limitations of the common light chain format. Then, we will describe the identification and optimization process for ERY974, an anti-glypican-3 × anti-CD3ε T cell-redirecting bispecific antibody with a common light chain. This format includes one of Chugai's proprietary technologies, termed ART-Ig technology, which consists of a method to identify a common light chain, isoelectric point (pI) engineering to purify the desired bispecific IgG antibody from byproducts, and Fc heterodimerization by an electrostatic steering effect. Furthermore, we describe some tips for de-risking the antibody when engineering a T cell redirecting antibody.


Subject(s)
Antibodies, Bispecific , Immunoglobulin G , Immunoglobulin Light Chains , Protein Engineering/methods , Animals , CD3 Complex/immunology , Glypicans/immunology , Humans , Mice
4.
Sci Transl Med ; 9(410)2017 Oct 04.
Article in English | MEDLINE | ID: mdl-28978751

ABSTRACT

Cancer care is being revolutionized by immunotherapies such as immune checkpoint inhibitors, engineered T cell transfer, and cell vaccines. The bispecific T cell-redirecting antibody (TRAB) is one such promising immunotherapy, which can redirect T cells to tumor cells by engaging CD3 on a T cell and an antigen on a tumor cell. Because T cells can be redirected to tumor cells regardless of the specificity of T cell receptors, TRAB is considered efficacious for less immunogenic tumors lacking enough neoantigens. Its clinical efficacy has been exemplified by blinatumomab, a bispecific T cell engager targeting CD19 and CD3, which has shown marked clinical responses against hematological malignancies. However, the success of TRAB in solid tumors has been hampered by the lack of a target molecule with sufficient tumor selectivity to avoid "on-target off-tumor" toxicity. Glypican 3 (GPC3) is a highly tumor-specific antigen that is expressed during fetal development but is strictly suppressed in normal adult tissues. We developed ERY974, a whole humanized immunoglobulin G-structured TRAB harboring a common light chain, which bispecifically binds to GPC3 and CD3. Using a mouse model with reconstituted human immune cells, we revealed that ERY974 is highly effective in killing various types of tumors that have GPC3 expression comparable to that in clinical tumors. ERY974 also induced a robust antitumor efficacy even against tumors with nonimmunogenic features, which are difficult to treat by inhibiting immune checkpoints such as PD-1 (programmed cell death protein-1) and CTLA-4 (cytotoxic T lymphocyte-associated protein-4). Immune monitoring revealed that ERY974 converted the poorly inflamed tumor microenvironment to a highly inflamed microenvironment. Toxicology studies in cynomolgus monkeys showed transient cytokine elevation, but this was manageable and reversible. No organ toxicity was evident. These data provide a rationale for clinical testing of ERY974 for the treatment of patients with GPC3-positive solid tumors.


Subject(s)
Antibodies, Bispecific/therapeutic use , Glypicans/immunology , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD3 Complex/metabolism , Cytokines/metabolism , Humans , Immunocompetence/drug effects , Injections, Intravenous , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macaca fascicularis , Mice, Transgenic , Steroids/pharmacology , Steroids/therapeutic use , T-Lymphocytes/drug effects
5.
PLoS One ; 11(6): e0156719, 2016.
Article in English | MEDLINE | ID: mdl-27258772

ABSTRACT

Semaphorin family proteins act on cells to mediate both repulsive and attractive guidance via binding to plexin family receptors, thereby playing fundamental roles in the morphogenesis and homeostasis of various tissues. Although semaphorin-plexin signaling is implicated in various diseases and is thus a target of intensive research, our mechanistic understanding of how semaphorins activate plexins on the cell surface is limited. Here, we describe unique anti-plexin-A1 antibodies that can induce a collapsed morphology in mouse dendritic cells as efficiently as the semaphorin 3A (Sema3A) ligand. Precise epitope analysis indicates that these "semaphorin-mimicking" antibodies dimerize cell-surface plexin-A1 by binding to the N-terminal sema domain of the plexin at sites away from the interface used by the Sema3A ligand. Structural analysis of plexin-A1 fragments using negative stain electron microscopy further revealed that this agonistic capacity is closely linked to the location and orientation of antibody binding. In addition, the full-length plexin-A1 ectodomain exhibited a highly curved "C" shape, reinforcing the very unusual dimeric receptor conformation of this protein at the cell surface when engaged with Sema3A or agonistic antibodies.


Subject(s)
Antibodies, Monoclonal/chemistry , Nerve Tissue Proteins/immunology , Receptors, Cell Surface/immunology , Semaphorin-3A/chemistry , Semaphorins/chemistry , Animals , Cell Membrane/metabolism , Dendritic Cells/cytology , Epitope Mapping , Epitopes , Humans , Mice , Nerve Tissue Proteins/chemistry , Protein Domains , Protein Multimerization , Receptors, Cell Surface/chemistry , Signal Transduction
6.
J Biol Chem ; 286(6): 4760-71, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21138846

ABSTRACT

The ectopic expression of transcription factors can reprogram differentiated tissue cells into induced pluripotent stem cells. However, this is a slow and inefficient process, depending on the simultaneous delivery of multiple genes encoding essential reprogramming factors and on their sustained expression in target cells. Moreover, once cell reprogramming is accomplished, these exogenous reprogramming factors should be replaced with their endogenous counterparts for establishing autoregulated pluripotency. Complete and designed removal of the exogenous genes from the reprogrammed cells would be an ideal option for satisfying this latter requisite as well as for minimizing the risk of malignant cell transformation. However, no single gene delivery/expression system has ever been equipped with these contradictory characteristics. Here we report the development of a novel replication-defective and persistent Sendai virus (SeVdp) vector based on a noncytopathic variant virus, which fulfills all of these requirements for cell reprogramming. The SeVdp vector could accommodate up to four exogenous genes, deliver them efficiently into various mammalian cells (including primary tissue cells and human hematopoietic stem cells) and express them stably in the cytoplasm at a prefixed balance. Furthermore, interfering with viral transcription/replication using siRNA could erase the genomic RNA of SeVdp vector from the target cells quickly and thoroughly. A SeVdp vector installed with Oct4/Sox2/Klf4/c-Myc could reprogram mouse primary fibroblasts quite efficiently; ∼1% of the cells were reprogrammed to Nanog-positive induced pluripotent stem cells without chromosomal gene integration. Thus, this SeVdp vector has potential as a tool for advanced cell reprogramming and for stem cell research.


Subject(s)
Cell Dedifferentiation/genetics , Fibroblasts , Genetic Vectors/genetics , Sendai virus/genetics , Stem Cells , Transduction, Genetic/methods , Animals , Cell Line , Humans , Kruppel-Like Factor 4 , Mice , Sendai virus/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
7.
Biosci Biotechnol Biochem ; 74(11): 2293-8, 2010.
Article in English | MEDLINE | ID: mdl-21071846

ABSTRACT

Sendai virus (SeV) is an enveloped virus with a non-segmented negative-strand RNA genome. SeV envelope fusion (F) glycoproteins play crucial roles in the viral life cycle in processes such as viral binding, assembly, and budding. In this study, we developed a viable recombinant SeV designated F-EGFP SeV/ΔF, in which the F protein was replaced by an F protein fused to EGFP at the carboxyl terminus. Living infected cells of the recombinant virus were directly visualized by green fluorescence. The addition of EGFP to the F protein maintained the activities of the F protein in terms of intracellular transport to the plasma membrane via the ER and the Golgi apparatus and fusion activity in the infected cells. These results suggest that this fluorescent SeV is a useful tool for studying the viral binding, assembly, and budding mechanisms of F proteins and the SeV life cycle in living infected cells.


Subject(s)
Green Fluorescent Proteins/genetics , Sendai virus/genetics , Viral Envelope Proteins/genetics , Fluorescence , Protein Transport , Recombinant Fusion Proteins , Research Design , Viral Fusion Proteins/physiology
8.
J Biol Chem ; 282(37): 27383-27391, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17623660

ABSTRACT

Persistent and stable expression of foreign genes has been achieved in mammalian cells by integrating the genes into the host chromosomes. However, this approach has several shortcomings in practical applications. For example, large scale production of protein pharmaceutics frequently requires laborious amplification of the inserted genes to optimize the gene expression. The random chromosomal insertion of exogenous DNA also results occasionally in malignant transformation of normal tissue cells, raising safety concerns in medical applications. Here we report a novel cytoplasmic RNA replicon capable of expressing installed genes stably without chromosome insertion. This system is based on the RNA genome of a noncytopathic variant Sendai virus strain, Cl.151. We found that this variant virus establishes stable symbiosis with host cells by escaping from retinoic acid-inducible gene I-interferon regulatory factor 3-mediated antiviral machinery. Using a cloned genome cDNA of Sendai virus Cl.151, we developed a recombinant RNA installed with exogenous marker genes that was maintained stably in the cytoplasm as a high copy replicon (about 4 x 10(4) copies/cell) without interfering with normal cellular function. Strong expression of the marker genes persisted for more than 6 months in various types of cultured cells and for at least two months in rat colonic mucosa without any apparent side effects. This stable RNA replicon is a potentially valuable genetic platform for various biological applications.


Subject(s)
Cytoplasm/metabolism , Replicon/genetics , Sendai virus/genetics , Animals , CHO Cells , Chickens , Cricetinae , Cricetulus , Genetic Vectors , Genome, Viral , Green Fluorescent Proteins/genetics , Interferon-beta/biosynthesis , Male , Phenotype , RNA, Viral/biosynthesis , Rats , Rats, Wistar , Signal Transduction
9.
J Biol Chem ; 280(3): 2028-35, 2005 Jan 21.
Article in English | MEDLINE | ID: mdl-15542612

ABSTRACT

Activated nucleotide sugars required for the synthesis of glycoconjugates within the secretory pathway of eukaryotes are provided by the action of nucleotide sugar transporters (NSTs). Typically, NSTs are studied in microsomal preparations from wild-type or mutant lines; however, in this setting it can be difficult to assess NST properties because of the presence of glycosyltransferases and other interfering activities. Here we have engineered Leishmania donovani to express high levels of an active LPG2 Golgi GDP-Man transporter bearing a C-terminal polyhistidine tag. The functional LPG2-HIS was solubilized, purified by metal affinity chromatography, and reconstituted into phosphatidylcholine-containing liposomes using polystyrene SM-2 beads. The proteoliposomes exhibited robust GDP-Man transport activity with an apparent K(m) of 6.6 mum. Transport activity was enhanced by preloading of GMP and showed specificity for multiple substrates (GDP-Ara and GDP-Fuc). In contrast to the activity in crude microsomes, transport was not dependent on the presence of divalent cations. Thus, reconstitution of transport activity using purified LPG2 protein in liposomes provides firm experimental evidence that a single polypeptide is solely required for NST activity and is able to mediate the uptake of multiple substrates. These studies are relevant to the study of NST structure and function in both protozoan parasites as well as their higher eukaryotic hosts.


Subject(s)
Glycosphingolipids/metabolism , Guanosine Diphosphate Mannose/metabolism , Leishmania donovani/metabolism , Membrane Proteins/metabolism , Protozoan Proteins/metabolism , Animals , Biological Transport , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Glycosphingolipids/isolation & purification , Liposomes , Membrane Proteins/isolation & purification , Protozoan Proteins/isolation & purification
10.
Infect Immun ; 72(6): 3622-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155672

ABSTRACT

Different Leishmania species rely to different extents on abundant glycoconjugates, such as lipophosphoglycan (LPG) and related molecules, in mammalian infections. Previously, we showed that Leishmania major deletion mutants lacking the Golgi GDP-mannose transporter LPG2, which is required for assembly of the dominant phosphoglycan (PG) repeats of LPG, were unable to survive in macrophages. These lpg2- mutants, however, retained the ability to generate asymptomatic, persistent infections in mice. In contrast, Ilg and colleagues showed that Leishmania mexicana LPG2 mutants retained virulence for mice. Here we identified a partial revertant population of the L. major lpg2- mutants (designated lpg2(-)REV) that had regained the ability to replicate in macrophages and induce disease pathology through a compensatory change. Like the lpg2 parent, the lpg2(-)REV revertant was unable to synthesize LPG2-dependent PGs in the promastigote stage and thus remained highly attenuated in the ability to induce infection. However, after considerable delay lpg2(-)REV revertant-infected mice exhibited lesions, and amastigotes isolated from these lesions were able to replicate within macrophages despite the fact that they were unable to synthesize PGs. Thus, in some respects, the lpg2(-)REV amastigotes resemble L. mexicana amastigotes. Future studies of the gene(s) responsible may shed light on the mechanisms employed by L. major to survive in the absence of LPG2-dependent glycoconjugates and may also improve the potential of the lpg2- L. major line to serve as a live parasite vaccine by overcoming its tendency to revert toward virulence.


Subject(s)
Glycosphingolipids/genetics , Leishmania major/growth & development , Leishmaniasis, Cutaneous/parasitology , Macrophages, Peritoneal/parasitology , Membrane Proteins/genetics , Mutation , Protozoan Proteins/genetics , Animals , Female , Glycoconjugates/metabolism , Glycosphingolipids/metabolism , Humans , Immunization , Leishmania major/genetics , Leishmania major/immunology , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/physiopathology , Leishmaniasis, Cutaneous/prevention & control , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Protozoan Proteins/metabolism , Protozoan Vaccines , Virulence
11.
Mol Biochem Parasitol ; 130(1): 31-42, 2003 Aug 11.
Article in English | MEDLINE | ID: mdl-14550894

ABSTRACT

Glycoconjugates have been shown to play important roles in Leishmania development. However, the ability to study these molecules and other processes would benefit greatly from improved methods for genetic manipulation and analysis of the amastigote stage. This is especially challenging for L. donovani, the agent of the most severe form of leishmaniasis, which can rapidly lose virulence during in vitro culture. Here we report on a clonal subline of an L. donovani 1S2D (LdBob or LdB), which differentiates readily from promastigotes to amastigotes in axenic culture, and maintains this ability during extended parasite cultivation in vitro. This derivative can be plated and transfected efficiently while grown as promastigotes or amastigotes. Importantly, LdB maintains the ability to differentiate while undergoing genetic alterations required for creation of gene knockouts and complemented lines. Like virulent L. donovani, LdB exhibits down-regulation of lipophosphoglycan (LPG) synthesis and up-regulation of A2 protein synthesis in amastigotes. We showed that knockouts of LPG2, encoding a Golgi GDP-mannose transporter, eliminated phosphoglycan synthesis in LdB axenic amastigotes. These and other data suggest that LdB axenic amastigotes will be generally useful as a differentiation model in studies of gene expression, virulence, glycoconjugate function and drug susceptibility in L. donovani.


Subject(s)
Glycosphingolipids/metabolism , Leishmania donovani/growth & development , Leishmania donovani/genetics , Animals , Antiprotozoal Agents/pharmacology , Culture Media , Gene Expression Regulation , Glycosphingolipids/genetics , Leishmania donovani/drug effects , Leishmania donovani/pathogenicity , Membrane Proteins/genetics , Membrane Proteins/metabolism , Parasitic Sensitivity Tests , Parasitology/methods , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Transfection , Virulence
12.
Science ; 301(5637): 1241-3, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12947201

ABSTRACT

Leishmania infections involve an acute phase of replication within macrophages, typically associated with pathology. After recovery parasites persist for long periods, which can lead to severe disease upon reactivation. Unlike the role of host factors, parasite factors affecting persistence are poorly understood. Leishmania major lacking phosphoglycans (lpg2-) were unable to survive in sand flies and macrophages, but retained the ability to persist indefinitely in the mammalian host without inducing disease. The L. major lpg2- thus provides a platform for probing parasite factors implicated in persistence and its role in disease and immunity.


Subject(s)
Glycosphingolipids/physiology , Leishmania major/physiology , Leishmaniasis, Cutaneous/parasitology , Macrophages/parasitology , Membrane Proteins/physiology , Phlebotomus/parasitology , Animals , Complement System Proteins/immunology , Cytokines/physiology , Female , Glycosphingolipids/genetics , Host-Parasite Interactions , Humans , Insect Vectors/parasitology , Leishmania major/genetics , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/pathology , Macrophage Activation , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Nitric Oxide/physiology , Phagosomes/parasitology , Protozoan Proteins/genetics , Protozoan Proteins/physiology , Virulence
13.
EMBO J ; 22(14): 3635-44, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12853478

ABSTRACT

Sulfation of all macromolecules entering the secretory pathway in higher organisms occurs in the Golgi and requires the high-energy sulfate donor adenosine 3'-phosphate 5'-phosphosulfate. Here we report the first molecular identification of a gene that encodes a transmembrane protein required to transport adenosine 3'-phosphate 5'-phosphosulfate from the cytosol into the Golgi lumen. Mutations in this gene, which we call slalom, display defects in Wg and Hh signaling, which are likely due to the lack of sulfation of glycosaminoglycans by the sulfotransferase sulfateless. Analysis of mosaic mutant ovaries shows that sll function is also essential for dorsal-ventral axis determination, suggesting that sll transports the sulfate donor required for sulfotransferase activity of the dorsal-ventral determinant pipe.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/embryology , Drosophila/genetics , Genes, Insect , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Amino Acid Sequence , Animals , Biological Transport , Body Patterning/genetics , Cytosol/metabolism , Drosophila Proteins/chemistry , Female , Gene Expression , Golgi Apparatus/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/chemistry , Molecular Sequence Data , Mutation , Ovary/metabolism , Proteoglycans/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Sulfate Transporters , Sulfates/metabolism , Wings, Animal/embryology
14.
Biosci Biotechnol Biochem ; 67(3): 592-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12723608

ABSTRACT

The roles of N-linked glycosylation in the intracellular transport and biological activities of the Sendai virus hemagglutinin-neuraminidase (HN) protein were studied. The protein contains four potential N-glycosylation sites: N77, N448, N499, and N511. By site-directed mutagenesis of these positions, the mature protein contained three N-linked oligosaccharides attached to N77, N499, and N511. The role of each added oligosaccharide in the structure and functions of the protein was identified by characterization of surface expression, hemadsorption, and neuraminidase activities of the corresponding mutant proteins. Elimination of the sites of N499 and N511 had the most detrimental effect, decreasing surface expression and hemadsorption. Elimination of the sites of N77 and N448 had similar but weaker effects. Mutants missing the sites of N499 and N511 were not able to induce syncytia formation in cells expressing mutant HN proteins and the fusion protein. Therefore, the N-linked oligosaccharides attached to N499 and N511 were important for intracellular transport and for the fusion promotion.


Subject(s)
HN Protein/chemistry , HN Protein/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Sendai virus/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antigens, Surface/analysis , Asparagine/genetics , Base Sequence , Binding Sites , COS Cells , Fluorescent Antibody Technique, Direct , Glycosylation , HN Protein/genetics , HeLa Cells , Hemadsorption , Humans , Mutagenesis, Site-Directed , Sendai virus/metabolism , Structure-Activity Relationship , Transfection , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
15.
FEBS Lett ; 513(2-3): 153-8, 2002 Feb 27.
Article in English | MEDLINE | ID: mdl-11904141

ABSTRACT

The selectivity and individual roles of the N-linked oligosaccharide chains of Sendai virus fusion protein (F protein) in the interaction with endoplasmic reticulum molecular chaperones were investigated by analyses of transient expression of single N-glycosylation mutants and sequential immunoprecipitation. We demonstrated differential interactions depending on the location of the N-linked oligosaccharide chain, and showed that these interactions were correlated with the folding and transport of F proteins. Moreover, mutant F proteins that lacked the specific N-linked oligosaccharide chains required for disulfide bond formation showed increased association with ERp57.


Subject(s)
Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Oligosaccharides/metabolism , Viral Fusion Proteins/metabolism , Calcium-Binding Proteins/metabolism , Calnexin , Gene Expression/drug effects , Glycosylation , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Indolizines/pharmacology , Isomerases/metabolism , Mutation , Precipitin Tests , Protein Disulfide-Isomerases , Protein Folding , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
16.
Eur J Biochem ; 269(1): 128-38, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11784306

ABSTRACT

A putative Drosophila nucleotide sugar transporter was characterized and shown to be the Drosophila homologue of the human UDP-Gal transporter (hUGT). When the Drosophila melanogaster UDP-Gal transporter (DmUGT) was expressed in mammalian cells, the transporter protein was localized in the Golgi membranes and complemented the UDP-Gal transport deficiency of Lec8 cells but not the CMP-Sia transport deficiency of Lec2 cells. DmUGT and hUGT were expressed in Saccharomyces cerevisiae cells in functionally active forms. Using microsomal vesicles isolated from Saccharomyces cerevisiae expressing these transporters, we unexpectedly found that both hUGT and DmUGT could transport UDP-GalNAc as well as UDP-Gal. When amino-acid residues that are conserved among human, murine, fission yeast and Drosophila UGTs, but are distinct from corresponding ones conserved among CMP-Sia transporters (CSTs), were substituted by those found in CST, the mutant transporters were still active in transporting UDP-Gal. One of these mutants in which Asn47 was substituted by Ala showed aberrant intracellular distribution with concomitant destabilization of the protein product. However, this mutation was suppressed by an Ile51 to Thr second-site mutation. Both residues were localized within the first transmembrane helix, suggesting that the structure of the helix contributes to the stabilization and substrate recognition of the UGT molecule.


Subject(s)
Monosaccharide Transport Proteins/physiology , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , Cloning, Molecular , Drosophila melanogaster , Humans , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Mutagenesis, Site-Directed , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...