Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 68: 167-79, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24798517

ABSTRACT

We compared the effects of three missense mutations in the GABAA receptor γ2 subunit on GABAA receptor assembly, trafficking and function in HEK293T cells cotransfected with α1, ß2, and wildtype or mutant γ2 subunits. The mutations R82Q and P83S were identified in families with genetic epilepsy with febrile seizures plus (GEFS+), and N79S was found in a single patient with generalized tonic-clonic seizures (GTCS). Although all three mutations were located in an N-terminal loop that contributes to the γ+/ß- subunit-subunit interface, we found that each mutation impaired GABAA receptor assembly to a different extent. The γ2(R82Q) and γ2(P83S) subunits had reduced α1ß2γ2 receptor surface expression due to impaired assembly into pentamers, endoplasmic reticulum (ER) retention and degradation. In contrast, γ2(N79S) subunits were efficiently assembled into GABAA receptors with only minimally altered receptor trafficking, suggesting that N79S was a rare or susceptibility variant rather than an epilepsy mutation. Increased structural variability at assembly motifs was predicted by R82Q and P83S, but not N79S, substitution, suggesting that R82Q and P83S substitutions were less tolerated. Membrane proteins with missense mutations that impair folding and assembly often can be "rescued" by decreased temperatures. We coexpressed wildtype or mutant γ2 subunits with α1 and ß2 subunits and found increased surface and total levels of both wildtype and mutant γ2 subunits after decreasing the incubation temperature to 30°C for 24h, suggesting that lower temperatures increased GABAA receptor stability. Thus epilepsy-associated mutations N79S, R82Q and P83S disrupted GABAA receptor assembly to different extents, an effect that could be potentially rescued by facilitating protein folding and assembly.


Subject(s)
Mutation, Missense/genetics , Protein Subunits/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Computer Simulation , Embryo, Mammalian , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Models, Molecular , Protein Subunits/genetics , Protein Transport/genetics , Rats , Receptors, GABA-A/drug effects , Temperature
2.
J Neurochem ; 111(6): 1501-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19811606

ABSTRACT

This is a study of the interaction between the two NMDA neurotransmitter receptor subtypes, NR1/NR2A and NR1/NR2B, and amyloid precursor protein (APP) 695, the major APP variant expressed in neurones. APP695 co-immunoprecipitated with assembled NR1-1a/NR2A and NR1-1a/NR2B NMDA receptors following expression in mammalian cells. Single NR1-1a, NR1-2a, NR1-4b(c-Myc), or NR2 subunit transfections revealed that co-association of APP695 with assembled NMDA receptors was mediated via the NR1 subunit; it was independent of the NR1 C1, C2, and C2' cassettes and, the use of an NR1-2a(c-Myc)-trafficking mutant suggested that interaction between the two proteins occurs in the endoplasmic reticulum. The use of antibodies directed against extracellular and intracellular NR2 subunit epitopes for immunoprecipitations suggested that APP/NMDA receptor association was mediated via N-terminal domains. Anti-APP antibodies immunoprecipitated NR1, NR2A, and NR2B immunoreactive bands from detergent extracts of mammalian brain; reciprocally, anti-NR1 or anti-NR2A antibodies co-immunoprecipitated APP immunoreactivity. Immune pellets from brain were sensitive to endoglycosidase H suggesting that, as for heterologous expression, APP and NMDA receptor association occurs in the endoplasmic reticulum. Co-expression of APP695 in mammalian cells resulted in enhanced cell surface expression of both NR1-1a/NR2A and NR1-1a/NR2B NMDA receptors with no increase in total subunit expression. These findings are further evidence for a role of APP in intracellular trafficking mechanisms. Further, they provide a link between two major brain proteins that have both been implicated in Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Amyloid beta-Protein Precursor/genetics , Cell Line, Transformed/ultrastructure , Cell Membrane/genetics , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Immunoprecipitation/methods , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/metabolism , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Protein Subunits/metabolism , Protein Transport/genetics , Protein Transport/physiology , Transfection/methods
3.
J Neurosci ; 29(2): 303-12, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19144831

ABSTRACT

Regulated trafficking of AMPA receptors to cell surface and to synapses is an important determinant of neuronal excitability. In the present study, we have addressed the role of agonist binding and desensitization in the early trafficking of glutamate receptor-D (GluR-D) AMPA receptors. Analysis of point-mutated GluR-D receptors, via electrophysiology and immunofluorescence, revealed that agonist-binding activity is essential for efficient delivery to cell surface in transfected cell lines and in neurons. Cotransfection with stargazin could fully rescue the surface expression of nonbinding mutant receptors in cell lines, indicating that stargazin is able to interact with and promote exit of AMPA receptors from endoplasmic reticulum (ER) independently of agonist binding. Secretion of separately expressed ligand-binding domain constructs showed a similar dependency of agonist binding to that observed with full-length GluR-D, supporting the idea that glutamate-induced closure of the binding site cleft is registered by ER quality control as a necessary priming step for transport competence. In contrast to agonist binding, the ability of the receptor to undergo desensitization had only a minor influence on trafficking. Our results are consistent with the hypothesis that AMPA receptors are synthesized as intrinsically unstable molecules, which require glutamate binding for structural stability and for transport-competence.


Subject(s)
Binding Sites/drug effects , Excitatory Amino Acid Agonists/pharmacology , Receptors, AMPA/metabolism , Animals , Binding Sites/genetics , Binding Sites/physiology , Biotinylation/methods , Cell Line, Transformed , Chlorocebus aethiops , Excitatory Amino Acid Antagonists/pharmacology , Green Fluorescent Proteins/genetics , Humans , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Molecular , Patch-Clamp Techniques/methods , Point Mutation/genetics , Protein Binding/drug effects , Protein Structure, Tertiary/genetics , Protein Transport/drug effects , Protein Transport/genetics , Quinoxalines/pharmacology , Receptors, AMPA/classification , Receptors, AMPA/genetics , Transfection/methods
4.
Blood ; 113(8): 1786-93, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19029439

ABSTRACT

Hemojuvelin (HJV) was recently identified as a critical regulator of iron homeostasis. It is either associated with cell membranes through a glycosylphosphatidylinositol anchor or released as a soluble form. Membrane-anchored HJV acts as a coreceptor for bone morphogenetic proteins and activates the transcription of hepcidin, a hormone that regulates iron efflux from cells. Soluble HJV antagonizes bone morphogenetic protein signaling and suppresses hepcidin expression. In this study, we examined the trafficking and processing of HJV. Cellular HJV reached the plasma membrane without obtaining complex oligosaccharides, indicating that HJV avoided Golgi processing. Secreted HJV, in contrast, has complex oligosaccharides and can be derived from HJV with high-mannose oligosaccharides at the plasma membrane. Our results support a model in which retrograde trafficking of HJV before cleavage is the predominant processing pathway. Release of HJV requires it to bind to the transmembrane receptor neogenin. Neogenin does not, however, play a role in HJV trafficking to the cell surface, suggesting that it could be involved either in retrograde trafficking of HJV or in cleavage leading to HJV release.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Golgi Apparatus/physiology , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Protein Transport/physiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Filipin/pharmacology , Furin/metabolism , GPI-Linked Proteins , Glycosylation , Hemochromatosis Protein , Humans , Liver Neoplasms/pathology , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Membrane Proteins/genetics , Oligosaccharides/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , RNA, Small Interfering
5.
Nephrol Dial Transplant ; 23(11): 3397-402, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18495742

ABSTRACT

BACKGROUND: Active Ca(2+) reabsorption in the kidney is facilitated by the epithelial transient receptor potential vanilloid Ca(2+) channel subtype 5 (TRPV5). The complex-glycosylated TRPV5 is expressed at the apical membrane of the renal distal convoluted tubule (DCT) cells where the pro-urine hormone klotho can stimulate its activity by N-oligosaccharide hydrolysis. This study investigates whether klotho and its closely related analogue, beta-glucuronidase, can activate other renal ion channels than TRPV5 expressed by DCT cells. METHODS: To determine the specificity of this stimulatory effect of klotho and beta-glucuronidase, a selection of ion channels and transporters expressed in the kidney (TRPV4, TRPV5, TRPV6 and TRPM6) was screened in transfected HEK293 cells by using Ca(2+)-influx measurements. RESULTS: Klotho and beta-glucuronidase have been found to significantly increase the activity of TRPV5 and TRPV6, but had no effect on TRPV4 and TRPM6. Furthermore, deglycosylation by endoglycosidase-F also stimulated the activity of TRPV4, TRPV5 and TRPV6, but not of TRPM6. CONCLUSIONS: These results suggest a modulating effect for klotho primarily restricted to the epithelial Ca(2+) channels TRPV5 and TRPV6.


Subject(s)
Calcium Channels/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glucuronidase/pharmacology , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , TRPV Cation Channels/metabolism , Calcium/metabolism , Calcium Channels/drug effects , Cell Line , Humans , Klotho Proteins , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , TRPV Cation Channels/drug effects , Transfection
6.
Pflugers Arch ; 454(1): 143-53, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17186307

ABSTRACT

Increase in mRNA expression and transport activity of the betaine gamma-amino-n-butyric acid cotransporter (BGAT) in response to hyperosmolality has been previously shown in MDCK cells. However, the hyperosmolality-induced response of endogenous BGAT protein expression was not investigated in detail. We show two forms of endogenous BGAT immunoreactivity that are expressed in MDCK II cells. Both are sensitive to Peptide N-Glycosidase F (PNGase F), suggesting that they are N-glycosylated proteins. One band, about 75 kDa, is resistant to Endo H, while the other 55 kDa band is sensitive to it, suggesting that they are fully N-glycosylated mature form in the post-Golgi compartment and core-glycosylated immature form in the endoplasmic reticulum (ER), respectively. When treated with hyperosmolality, they are significantly increased. But the rate of BGAT processing, as assessed by the ratio of mature to immature form, is not increased, suggesting that hyperosmolality does not facilitate the export of BGAT from the ER to the secretory pathway. Surface biotinylation and confocal microscopy show that hyperosmolality significantly increases the amount of the mature form of BGAT on the basolateral membrane with a very small fraction on the apical membrane. We conclude that BGAT is an N-glycosylated protein with two glycoforms and endogenous BGAT synthesis rather than processing is involved in the adaptation to the hyperosmotic stress.


Subject(s)
GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/metabolism , Kidney/metabolism , Animals , Blotting, Western , Cell Line , Cell Membrane/metabolism , Dogs , GABA Plasma Membrane Transport Proteins/drug effects , Glycosylation , Immunohistochemistry , Intracellular Membranes/metabolism , Kidney/cytology , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Osmotic Pressure , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Protein Isoforms/drug effects , Protein Processing, Post-Translational , Time Factors
7.
Article in English | MEDLINE | ID: mdl-16945565

ABSTRACT

A novel endogenous beta-1,4-endoglucanase (Ag-EGase III) gene belonging to the glycoside hydrolase family (GHF) 5 was cloned from the mulberry longicorn beetle, Apriona germari. The Ag-EGase III gene spans 1061 bp and consists of a single exon coding for 325 amino acid residues. The Ag-EGase III showed 89% protein sequence identity to another beetle, Psacothea hilaris, cellulase belonging to GHF 5. The Ag-EGase III has the potential proton donor and nucleophile amino acids conserved in GHF 5 and two putative N-glycosylation sites. Northern blot and Western blot analyses showed that Ag-EGases were expressed in the gut; Ag-EGase III and Ag-EGase I were expressed in three gut regions, and no Ag-EGase II was found in hindgut, indicating that the foregut and midgut are the prime sites for cellulase synthesis in A. germari larvae. The cDNA encoding Ag-EGase III was expressed as a 47-kDa polypeptide in baculovirus-infected insect Sf9 cells and the enzyme activity of the purified recombinant Ag-EGase III was approximately 1037 U per mg of recombinant Ag-EGase III. The enzymatic property of the purified recombinant Ag-EGase III showed the highest activity at 55 degrees C and pH 6.0, and was stable at 60 degrees C at least for 10 min. In addition, the N-glycosylation of Ag-EGase III was revealed by treatment with tunicamycin of recombinant virus-infected insect Sf9 cells and with endoglycosidase F of purified recombinant Ag-EGase III, demonstrating that the carbohydrate moieties are not necessary for enzyme activity.


Subject(s)
Cellulase/genetics , Coleoptera/enzymology , Animals , Cellulase/metabolism , Cloning, Molecular , Coleoptera/genetics , Gene Expression Regulation, Enzymologic , Glycosylation/drug effects , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Morus , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Species Specificity , Tunicamycin/pharmacology
8.
FEBS J ; 273(14): 3287-300, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16792699

ABSTRACT

N-Glycosylation is a cotranslational and post-translational process of proteins that may influence protein folding, maturation, stability, trafficking, and consequently cell surface expression of functional channels. Here we have characterized two consensus N-glycosylation sequences of a voltage-gated K+ channel (Kv3.1). Glycosylation of Kv3.1 protein from rat brain and infected Sf9 cells was demonstrated by an electrophoretic mobility shift assay. Digestion of total brain membranes with peptide N glycosidase F (PNGase F) produced a much faster-migrating Kv3.1 immunoband than that of undigested brain membranes. To demonstrate N-glycosylation of wild-type Kv3.1 in Sf9 cells, cells were treated with tunicamycin. Also, partially purified proteins were digested with either PNGase F or endoglycosidase H. Attachment of simple-type oligosaccharides at positions 220 and 229 was directly shown by single (N229Q and N220Q) and double (N220Q/N229Q) Kv3.1 mutants. Functional measurements and membrane fractionation of infected Sf9 cells showed that unglycosylated Kv3.1s were transported to the plasma membrane. Unitary conductance of N220Q/N229Q was similar to that of the wild-type Kv3.1. However, whole cell currents of N220Q/N229Q channels had slower activation rates, and a slight positive shift in voltage dependence compared to wild-type Kv3.1. The voltage dependence of channel activation for N229Q and N220Q was much like that for N220Q/N229Q. These results demonstrate that the S1-S2 linker is topologically extracellular, and that N-glycosylation influences the opening of the voltage-dependent gate of Kv3.1. We suggest that occupancy of the sites is critical for folding and maturation of the functional Kv3.1 at the cell surface.


Subject(s)
Consensus Sequence , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Brain/cytology , Cell Fractionation , Cell Membrane/chemistry , Cell Membrane/metabolism , Cells, Cultured , Conserved Sequence , Glycosylation , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Membrane Proteins/genetics , Models, Biological , Molecular Sequence Data , Mutation , Patch-Clamp Techniques , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Rats , Sequence Homology, Amino Acid , Shaw Potassium Channels/genetics , Spodoptera/cytology , Spodoptera/drug effects , Tunicamycin/pharmacology
9.
J Virol ; 80(8): 3994-4004, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16571816

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) glycoprotein 5 (GP5) is the most abundant envelope glycoprotein and a major inducer of neutralizing antibodies in vivo. Three putative N-linked glycosylation sites (N34, N44, and N51) are located on the GP5 ectodomain, where a major neutralization epitope also exists. To determine which of these putative sites are used for glycosylation and the role of the glycan moieties in the neutralizing antibody response, we generated a panel of GP5 mutants containing amino acid substitutions at these sites. Biochemical studies with expressed wild-type (wt) and mutant proteins revealed that the mature GP5 contains high-mannose-type sugar moieties at all three sites. These mutations were subsequently incorporated into a full-length cDNA clone. Our data demonstrate that mutations involving residue N44 did not result in infectious progeny production, indicating that N44 is the most critical amino acid residue for infectivity. Viruses carrying mutations at N34, N51, and N34/51 grew to lower titers than the wt PRRSV. In serum neutralization assays, the mutant viruses exhibited enhanced sensitivity to neutralization by wt PRRSV-specific antibodies. Furthermore, inoculation of pigs with the mutant viruses induced significantly higher levels of neutralizing antibodies against the mutant as well as the wt PRRSV, suggesting that the loss of glycan residues in the ectodomain of GP5 enhances both the sensitivity of these viruses to in vitro neutralization and the immunogenicity of the nearby neutralization epitope. These results should have great significance for development of PRRSV vaccines of enhanced protective efficacy.


Subject(s)
Antibodies, Viral/blood , Porcine respiratory and reproductive syndrome virus/immunology , Porcine respiratory and reproductive syndrome virus/physiology , Viral Envelope Proteins/metabolism , Animals , Cell Line , Cricetinae , Epitopes , Glycosylation , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Mutation , Swine , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
10.
J Virol ; 79(10): 6152-61, 2005 May.
Article in English | MEDLINE | ID: mdl-15858000

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV), a member of the genus Nairovirus of the family Bunyaviridae, causes severe disease with high rates of mortality in humans. The CCHFV M RNA segment encodes the virus glycoproteins G(N) and G(C). To understand the processing and intracellular localization of the CCHFV glycoproteins as well as their neutralization and protection determinants, we produced and characterized monoclonal antibodies (MAbs) specific for both G(N) and G(C). Using these MAbs, we found that G(N) predominantly colocalized with a Golgi marker when expressed alone or with G(C), while G(C) was transported to the Golgi apparatus only in the presence of G(N). Both proteins remained endo-beta-N-acetylglucosaminidase H sensitive, indicating that the CCHFV glycoproteins are most likely targeted to the cis Golgi apparatus. Golgi targeting information partly resides within the G(N) ectodomain, because a soluble version of G(N) lacking its transmembrane and cytoplasmic domains also localized to the Golgi apparatus. Coexpression of soluble versions of G(N) and G(C) also resulted in localization of soluble G(C) to the Golgi apparatus, indicating that the ectodomains of these proteins are sufficient for the interactions needed for Golgi targeting. Finally, the mucin-like and P35 domains, located at the N terminus of the G(N) precursor protein and removed posttranslationally by endoproteolysis, were required for Golgi targeting of G(N) when it was expressed alone but were dispensable when G(C) was coexpressed. In neutralization assays on SW-13 cells, MAbs to G(C), but not to G(N), prevented CCHFV infection. However, only a subset of G(C) MAbs protected mice in passive-immunization experiments, while some nonneutralizing G(N) MAbs efficiently protected animals from a lethal CCHFV challenge. Thus, neutralization of CCHFV likely depends not only on the properties of the antibody, but on host cell factors as well. In addition, nonneutralizing antibody-dependent mechanisms, such as antibody-dependent cell-mediated cytotoxicity, may be involved in the in vivo protection seen with the MAbs to G(C).


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Glycoproteins/immunology , Glycoproteins/metabolism , Golgi Apparatus/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever, Crimean/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Line , Disease Models, Animal , Hemorrhagic Fever, Crimean/prevention & control , Humans , Immunization, Passive , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Mice , Mice, Inbred BALB C , Neutralization Tests , Solubility
11.
J Neurosci ; 25(9): 2215-25, 2005 Mar 02.
Article in English | MEDLINE | ID: mdl-15745947

ABSTRACT

The glutamate receptor (GluR) agonist-binding site consists of amino acid residues in the extracellular S1 and S2 domains in the N-terminal and M3-M4 loop regions, respectively. In the present study, we sought to confirm that the conserved ligand-binding residues identified in the AMPA receptor S1S2 domains also participate in ligand binding of GluR6 kainate receptors. Amino acid substitutions were made in the GluR6 parent at R523, T690, and E738 to alter their potential interactions with ligand. Mutant receptors were expressed in human embryonic kidney 293 cells, confirmed by Western blot analysis, and tested by [3H]kainate binding and patch-clamp recording. Each of the binding site mutations was sufficient to reduce [3H]kainate binding to undetectable levels and eliminate functional responses to glutamate or kainate. As with our studies of other nonfunctional mutants (Fleck et al., 2003), immunocytochemical staining and cell-surface biotinylation studies showed that the mutant receptors were retained intracellularly and did not traffic to the cell surface. Endoglycosidase-H digests and colocalization with endoplasmic reticulum (ER) markers demonstrated that the mutant receptors are immaturely glycosylated and retained in the ER. Immunoprecipitation, native PAGE, and functional studies confirmed that the GluR6-binding site mutants are capable of multimeric assembly, indicating their retention in the ER does not result from a gross protein folding error. Together, these results confirm the role of R523, T690, and E738 directly in ligand binding to GluR6 and further support our previous report that nonfunctional GluRs are retained intracellularly by a functional checkpoint in ER quality control.


Subject(s)
Endoplasmic Reticulum/physiology , Ligands , Protein Transport/physiology , Receptors, Kainic Acid/metabolism , Amino Acid Substitution/physiology , Binding Sites/physiology , Biotinylation/methods , Blotting, Western/methods , Cell Line , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Gene Expression/genetics , Glutamic Acid/pharmacology , Glycosylation/drug effects , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Kainic Acid/pharmacokinetics , Luminescent Proteins , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Membrane Potentials/genetics , Membrane Potentials/radiation effects , Microscopy, Confocal/methods , Models, Molecular , Mutagenesis, Site-Directed/methods , Mutation/physiology , Patch-Clamp Techniques/methods , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Protein Binding/genetics , Protein Binding/physiology , Radioligand Assay/methods , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/genetics , Sequence Alignment/methods , Structure-Activity Relationship , Transfection/methods , Tritium/pharmacokinetics , GluK2 Kainate Receptor
12.
J Biol Chem ; 280(15): 15173-9, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15708861

ABSTRACT

pFGE is the paralog of the formylglycine-generating enzyme (FGE), which catalyzes the oxidation of a specific cysteine to Calpha-formylglycine, the catalytic residue in the active site of sulfatases. The enzymatic activity of sulfatases depends on this posttranslational modification, and the genetic defect of FGE causes multiple sulfatase deficiency. The structural and functional properties of pFGE were analyzed. The comparison with FGE demonstrates that both share a tissue-specific expression pattern and the localization in the lumen of the endoplasmic reticulum. Both are retained in the endoplasmic reticulum by a saturable mechanism. Limited proteolytic cleavage at similar sites indicates that both also share a similar three-dimensional structure. pFGE, however, is lacking the formylglycine-generating activity of FGE. Although overexpression of FGE stimulates the generation of catalytically active sulfatases, overexpression of pFGE has an inhibitory effect. In vitro pFGE interacts with sulfatase-derived peptides but not with FGE. The inhibitory effect of pFGE on the generation of active sulfatases may therefore be caused by a competition of pFGE and FGE for newly synthesized sulfatase polypeptides.


Subject(s)
Alanine/analogs & derivatives , Glycine/analogs & derivatives , Sulfatases/biosynthesis , Sulfatases/chemistry , Alanine/chemistry , Binding Sites , Binding, Competitive , Blotting, Northern , Blotting, Western , Catalysis , Cell Line , Codon , Cross-Linking Reagents/pharmacology , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Glycine/chemistry , Glycosylation , Humans , Immunoprecipitation , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Mass Spectrometry , Microscopy, Immunoelectron , Molecular Sequence Data , Oxidoreductases Acting on Sulfur Group Donors , Peptides/chemistry , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Skin/metabolism , Time Factors , Tissue Distribution , Transfection , Trypsin/chemistry , Two-Hybrid System Techniques
13.
J Biol Chem ; 279(45): 46755-63, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15337749

ABSTRACT

Reduced folates such as 5-methyl tetrahydrofolate and classical antifolates such as methotrexate are actively transported into mammalian cells by the reduced folate carrier (RFC). RFC is characterized by 12 stretches of mostly hydrophobic, alpha-helix-promoting amino acids, internally oriented N and C termini, and a large central linker connecting transmembrane domains (TMDs) 1-6 and 7-12. Previous studies showed that deletion of the majority of the central loop domain between TMDs 6 and 7 abolished transport, but this segment could be replaced with mostly non-homologous sequence from the SLC19A2 thiamine transporter to restore transport function. In this report, we expressed RFC from separate TMD1-6 and TMD7-12 RFC half-molecule constructs, each with a unique epitope tag, in RFC-null K562 cells to restore transport activity. Restored transport exhibited characteristic transport kinetics for methotrexate, a capacity for trans-stimulation by pretreatment with leucovorin, and inhibition by N-hydroxysuccinimide methotrexate, a documented affinity inhibitor of RFC. The TMD1-6 half-molecule migrated on SDS gels as a 38-58 kDa glycosylated species and was converted to 27 kDa by N-glycosidase F or tunicamycin treatments. The 40 kDa TMD7-12 half-molecule was unaffected by these treatments. Using transfected cells expressing both TMDs 1-6 and TMDs 7-12 as separate polypeptides, the TMD7-12 half-molecule was covalently radiolabeled with N-hydroxysuccinimide [(3)H]methotrexate. No radioactivity was incorporated into the TMD1-6 half-molecule. Digestion with endoproteinase GluC decreased the size of the radiolabeled 40 kDa TMD7-12 polypeptide to approximately 20 kDa. Our results demonstrate that a functional RFC can be reconstituted with RFC half-molecules and localize a critical substrate binding domain to within TMDs 7-12.


Subject(s)
Membrane Transport Proteins/metabolism , Methotrexate/analogs & derivatives , Amino Acid Sequence , Biological Transport , Cell Line , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Epitopes/chemistry , Folic Acid/chemistry , Folic Acid Antagonists/pharmacology , Glycosylation , Humans , Immunoprecipitation , K562 Cells , Kinetics , Leucovorin/pharmacology , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Methotrexate/pharmacology , Microscopy, Confocal , Molecular Sequence Data , Peptides/chemistry , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Reduced Folate Carrier Protein , Sodium Dodecyl Sulfate/chemistry , Substrate Specificity , Thiamine/chemistry , Time Factors , Transfection , Tunicamycin/pharmacology
14.
AIDS Res Hum Retroviruses ; 18(17): 1311-7, 2002 Nov 20.
Article in English | MEDLINE | ID: mdl-12487819

ABSTRACT

Mannose-binding lectin (MBL) is a C-type lectin of the innate immune system that binds to carbohydrates on the surface of certain microorganisms. Previous studies showed that MBL binds to gp120, the envelope glycoprotein of HIV-1. gp120 is extensively glycosylated, with N-linked complex and high mannose carbohydrates accounting for about half of the molecular weight. The objectives of this study were to determine the types of glycans on gp120 important for MBL binding and to determine if alteration of complex glycans with neuraminidase (NA) could enhance the interaction of MBL with virus. Lectin blot analyses revealed that MBL interacted with recombinant gp120 (rgp120) from both T cell-tropic and M-tropic virus strains. Treatment of rgp120 with endoglycosidase H (eH) or endoglycosidase F1 (eF1) abrogated binding of MBL, but did not decrease binding of wheat germ agglutinin indicating that high mannose and/or hybrid N-linked glycans were required for MBL binding. Removal of sialic acids from rgp120 with NA enhanced MBL binding. Treatment of intact virus from T cell lines or primary isolates with eF1 also significantly decreased HIV binding to MBL, while treatment with NA substantially increased binding. Treatment of virus with both eF1 and NA did not decrease binding compared to NA alone suggesting that NA treatment exposed binding sites on gp120 that are not high mannose glycans. These studies provide evidence that MBL binds to HIV via high mannose carbohydrates on gp120 and shows that the interaction of MBL with virus is regulated by sialylation.


Subject(s)
HIV Envelope Protein gp120/metabolism , Mannose-Binding Lectin/metabolism , Mannose/physiology , N-Acetylneuraminic Acid/physiology , Polysaccharides/physiology , HIV Envelope Protein gp120/chemistry , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Neuraminidase/pharmacology
15.
Arch Biochem Biophys ; 392(2): 279-86, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11488603

ABSTRACT

The white-rot fungus Phellinus ribis produced a single form of laccase, which was purified to apparent electrophoretic homogeneity from cultures induced with 2,5-xylidine. This protein was a dimer, consisting of two subunits of 76 kDa as determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Carbohydrate analysis revealed that the enzyme contained about 28% carbohydrate content. The laccase appeared to be different from other known laccases by the UV-visible absorption spectrum analysis. One enzyme molecule contained one copper, one manganese, and two zinc atoms. The laccase showed optimal activity at pH 4.0-6.0, 5.0, and 6.0 with 2,6-dimethoxyphenol, ABTS [2,2'-azino-bis-(3-ethylbenzothiazoline-6-sulfonic acid)], and syringaldazine, respectively. The enzyme preferably oxidized dimethoxyphenol and aromatic amine compounds. The stability of the laccase was low at acidic pH, whereas it showed high stability at neutral pH and mild temperature. The N-terminal amino acid sequence revealed a very low homology with other microbial laccases. With some substrates, the addition of manganese and H2O2 resulted in a remarkable increase in the oxidation rate. Without an appropriate phenolic substrate, the enzyme could not oxidize Mn(II) in the presence of H2O2 or pyrophosphate.


Subject(s)
Basidiomycota/enzymology , Fungal Proteins/chemistry , Oxidoreductases/chemistry , Amino Acid Sequence , Benzothiazoles , Carbohydrates/chemistry , Catalysis , Dimerization , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Hydrazones/pharmacology , Hydrogen Peroxide/pharmacology , Hydrogen-Ion Concentration , Laccase , Lectins/metabolism , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Molecular Sequence Data , Oxygen/metabolism , Pyrogallol/analogs & derivatives , Pyrogallol/pharmacology , Sequence Homology, Amino Acid , Spectrophotometry , Sulfonic Acids/pharmacology , Temperature , Time Factors , Ultraviolet Rays
16.
Comp Biochem Physiol B Biochem Mol Biol ; 129(4): 863-70, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435141

ABSTRACT

Neutral endopeptidase (NEP) is a zinc metallopeptidase ubiquitously distributed in various tissues in mammals. This peptidase is involved in the post-secretory metabolism of various neuropeptides and peptide hormones in vivo, such as enkephalins, bradykinin, atrial natriuretic peptide, substance P and endothelins. In this paper we show that NEP is expressed in ovaries as a 110-kDa glycosylated integral membrane protein with enzymatic properties similar to those of the kidney protein. Using immunohistochemistry, we localize the peptidase in the granulosa cells of follicles at all stages of maturation, with the exception of atretic follicles. We also observe immunoreactive staining in the epithelia that lines the blood vessels in the medulla and the surface of the ovary. The co-localization of NEP and bioactive peptides known to be physiological substrates of NEP in other tissues suggests an important role for this protein in processes such as follicle maturation, ovulation, and/or regulation of ovarian blood flow, by modulating the physiological function of these peptides.


Subject(s)
Granulosa Cells/enzymology , Neprilysin/biosynthesis , Ovary/enzymology , Animals , Blotting, Western , Cell Membrane/chemistry , Epithelial Cells/enzymology , Female , Immunoblotting , Immunohistochemistry , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Peptides/chemistry , Rabbits , Time Factors
17.
J Biol Chem ; 276(30): 27981-8, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11375981

ABSTRACT

The rat transporter rCNT1 is the archetype of a family of concentrative nucleoside transporters (CNTs) found both in eukaryotes and in prokaryotes. In the present study we have used antibodies to investigate the subcellular distribution and membrane topology of this protein. rCNT1 was found to be expressed predominantly in the brush-border membranes of the polarized epithelial cells of rat jejunum and renal cortical tubules and in the bile canalicular membranes of liver parenchymal cells, consistent with roles in the absorption of dietary nucleosides, of nucleosides in the glomerular filtrate, or of nucleosides arising from the action of extracellular nucleotidases, respectively. The effect of endoglycosidase F treatment on wild-type and mutant rCNT1 expressed in Xenopus oocytes revealed that the recombinant transporter could be glycosylated at either or both of Asn605 and Asn643, indicating that its C terminus is extracellular. In contrast, potential N-glycosylation sites introduced near the N terminus, or between putative transmembrane (TM) helices 4 and 5, were not glycosylated. The deduced orientation of the N terminus in the cytoplasm was confirmed by immunocytochemistry on intact and saponin-permeabilized Chinese hamster ovary cells expressing recombinant rCNT1. These results, in conjunction with extensive analyses of CNT family protein sequences using predictive algorithms, lead us to propose a revised topological model, in which rCNT1 possesses 13 TM helices with the hydrophilic N-terminal and C-terminal domains on the cytoplasmic and extracellular sides of the membrane, respectively. Furthermore, we show that the first three TM helices, which are absent from prokaryote CNTs, are not essential for transporter function; truncated proteins lacking these helices, derived either from rCNT1 or from its human homolog hCNT1, were found to retain significant sodium-dependent uridine transport activity when expressed in oocytes.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/physiology , Membrane Transport Proteins , Amino Acid Motifs , Animals , Asparagine/chemistry , Biological Transport , Blotting, Western , CHO Cells , Cell Membrane/metabolism , Cricetinae , DNA, Complementary/metabolism , Gene Deletion , Glycosylation , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Kidney/metabolism , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Mutagenesis, Site-Directed , Mutation , Oocytes/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Recombinant Proteins/metabolism , Tissue Distribution , Transcription, Genetic , Transfection , Uridine/metabolism , Xenopus
18.
Lipids ; 35(5): 495-502, 2000 May.
Article in English | MEDLINE | ID: mdl-10907784

ABSTRACT

An extracellular 1,3-specific lipase with molecular weight of 35.5 kDa and an isoelectric point of 4.4 from Aspergillus niger has been purified 50-fold by pH precipitation followed by a series of chromatographic steps with an overall yield of 10%. The enzyme was homogeneous as judged by denaturing polyacrylamide gel electrophoresis and size-exclusion fast-performance liquid chromatography. It contained 2.8% sugar which was completely removed by endoglycosidase F treatment, and the deglycosylated enzyme retained full activity. The native lipase showed optimal activity between temperatures 35 and 55 degrees C and pH 5.0 and 6.0. The amino acid composition and the N-terminal sequence were found to be different from lipases previously purified from A. niger. The enzyme was resistant to trypsin, chymotrypsin, endoprotease Glu-C, thrombin, and papain under native conditions but was susceptible to cleavage by the same proteases when heat-denatured.


Subject(s)
Aspergillus niger/enzymology , Lipase/chemistry , Lipase/isolation & purification , Chromatography , Chromatography, Liquid , Chromatography, Thin Layer , Chymotrypsin/pharmacology , Electrophoresis, Polyacrylamide Gel , Hydrogen-Ion Concentration , Isoelectric Focusing , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Papain/pharmacology , Temperature , Thrombin/pharmacology , Time Factors , Trypsin/pharmacology
19.
FEBS Lett ; 475(2): 117-20, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10858500

ABSTRACT

Aspergillus nidulans catalase B (CatB) was purified to homogeneity and characterized as a hydroperoxidase which resembles typical catalases in some physicochemical characteristics: (1) it has an apparent molecular weight of 360000 and is composed of four glycosylated subunits, (2) it has hydrophobic properties as revealed by extractability in ethanol/chloroform and binding to phenyl-Superose, and (3) it has an acidic isoelectric point at pH 3. 5. Also CatB exhibits some distinctive properties, e.g. it is not inhibited by the presence of 2% sodium dodecyl sulfate, 9 M urea or reducing agents. Furthermore, even though CatB does not exhibit any residual peroxidase activity, it is able to retain up to 38% of its initial catalase activity after incubation with the typical catalase inhibitor 3-amino-1,2,4-triazole.


Subject(s)
Aspergillus nidulans/enzymology , Catalase/chemistry , Amitrole/pharmacology , Anti-Bacterial Agents/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Enzyme Stability , Glycosylation , Hydrogen-Ion Concentration , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Peroxidases/metabolism , Reducing Agents/pharmacology , Sodium Dodecyl Sulfate/pharmacology , Surface-Active Agents/pharmacology , Time Factors , Tunicamycin/pharmacology , Urea/pharmacology
20.
Curr Biol ; 9(16): 927-30, 1999 Aug 26.
Article in English | MEDLINE | ID: mdl-10469599

ABSTRACT

Inhibitory immunoreceptors downregulate signaling by recruiting Src homology 2 (SH2) domain-containing tyrosine and/or lipid phosphatases to activating receptor complexes [1]. There are indications that some inhibitory receptors might also perform other functions [2] [3]. In adherent macrophages, two inhibitory receptors, SHPS-1 and PIR-B, are the major proteins binding to the tyrosine phosphatase SHP-1. SHPS-1 also associates with two tyrosine-phosphorylated proteins (pp55 and pp130) and a protein tyrosine kinase [4]. Here, we have identified pp55 and pp130 as the adaptor molecules SKAP55hom/R (Src-kinase-associated protein of 55 kDa homologue) and FYB/SLAP-130 (Fyn-binding protein/SLP-76-associated protein of 130 kDa), respectively, and the tyrosine kinase activity as PYK2. Two distinct SHPS-1 complexes were formed, one containing SKAP55hom/R and FYB/SLAP-130, and the other containing PYK2. Recruitment of FYB/SLAP-130 to SHPS-1 required SKAP55hom/R, whereas PYK2 associated with SHPS-1 independently. Formation of both complexes was independent of SHP-1 and tyrosine phosphorylation of SHPS-1. Finally, tyrosine phosphorylation of members of the SHPS-1 complexes was regulated by integrin-mediated adhesion. Thus, SHPS-1 provides a scaffold for the assembly of multi-protein complexes that might both transmit adhesion-regulated signals and help terminate such signals through SHP-1-directed dephosphorylation. Other inhibitory immunoreceptors might have similar scaffold-like functions.


Subject(s)
Antigens, Differentiation , Bone Marrow Cells/chemistry , Macrophages/chemistry , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Protein Folding , Receptors, Immunologic , Animals , COS Cells , Cell Adhesion , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Immunoblotting , Macrophages/drug effects , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Mice , Mice, Mutant Strains , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/metabolism , Sulfones/analysis , Sulfones/metabolism , Uridine/analogs & derivatives , Uridine/analysis , Uridine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...