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










Publication year range
1.
Curr Res Transl Med ; 71(2): 103380, 2023.
Article in English | MEDLINE | ID: mdl-36738659

ABSTRACT

PURPOSE OF THE STUDY: Calreticulin is an endoplasmic reticulum chaperone protein, which is involved in protein folding and in peptide loading of major histocompatibility complex class I molecules together with its homolog calnexin. Mutated calreticulin is associated with a group of hemopoietic disorders, especially myeloproliferative neoplasms. Currently only the cellular immune response to mutated calreticulin has been described, although preliminary findings have indicated that antibodies to mutated calreticulin are not specific for myeloproliferative disorders. These findings have prompted us to characterize the humoral immune response to mutated calreticulin and its chaperone homologue calnexin. PATIENTS AND METHODS: We analyzed sera from myeloproliferative neoplasm patients, healthy donors and relapsing-remitting multiple sclerosis patients for the occurrence of autoantibodies to wild type and mutated calreticulin forms and to calnexin by enzyme-linked immunosorbent assay. RESULTS: Antibodies to mutated calreticulin and calnexin were present at similar levels in serum samples of myeloproliferative neoplasm and multiple sclerosis patients as well as healthy donors. Moreover, a high correlation between antibodies to mutated calreticulin and calnexin was seen for all patient and control groups. Epitope binding studies indicated that cross-reactive antibodies bound to a three-dimensional epitope encompassing a short linear sequence in the C-terminal of mutated calreticulin and calnexin. CONCLUSION: Collectively, these findings indicate that calreticulin mutations may be common and not necessarily lead to onset of myeloproliferative neoplasm, possibly due to elimination of cells with mutations. This, in turn, may suggest that additional molecular changes may be required for development of myeloproliferative neoplasm.


Subject(s)
Calreticulin , Neoplasms , Humans , Calreticulin/genetics , Calnexin/genetics , Calnexin/chemistry , Calnexin/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
2.
Insect Biochem Mol Biol ; 41(2): 109-17, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21078390

ABSTRACT

Antifreeze proteins (AFPs) are characterized by their capacity to inhibit the growth of ice and are produced by a variety of polar fish, terrestrial arthropods and other organisms inhabiting cold environments. This capacity reflects their role as stabilizers of supercooled body fluids. The longhorn beetle Rhagium inquisitor is known to express AFPs in its body fluids. In this work we report on the primary structure and structural characteristics of a 12.8 kDa AFP from this beetle (RiAFP). It has a high capacity to evoke antifreeze activity as compared to other known insect AFPs and it is structurally unique in several aspects. In contrast to the high content of disulfide bond-formation observed in other coleopteran AFPs, RiAFP contains only a single such bond. Six internal repeat segments of a thirteen residue repeat pattern is irregularly spaced apart throughout its sequence. The central part of these repeat segments is preserved as TxTxTxT, which is effectively an expansion of the TxT ice-binding motif found in the AFPs of several known insect AFPs.


Subject(s)
Antifreeze Proteins/metabolism , Insect Proteins/metabolism , Amino Acid Sequence , Animals , Antifreeze Proteins/genetics , Cold Temperature , Coleoptera/genetics , Coleoptera/metabolism , Freezing , Hemolymph/physiology , Ice , Insect Proteins/genetics , Larva/genetics , Larva/metabolism , Models, Molecular , Molecular Sequence Data , Protein Folding , Sequence Analysis
3.
Protein Pept Lett ; 17(1): 121-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20214635

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit tumor growth and angiogenesis. Covalent linkage of naproxen to human serum albumin (HSA) has been shown to target it efficiently to the liver and this may potentially be exploited for liver-selective inhibition of angiogenesis. With the aim of investigating the anti-angiogenic efficiency of NSAID-HSA conjugates in vitro, three NSAIDs, aspirin, ibuprofen, and naproxen were conjugated to HSA using different concentrations of their N-hydroxysuccinimide esters. Conjugation ratios from 10 to 50 were achieved and the conjugates retained a growth inhibitory effect on endothelial cells at or above the level of the non-conjugated NSAIDs in an in vitro angiogenesis assay.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Serum Albumin/chemistry , Serum Albumin/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Coculture Techniques , Endothelial Cells , Fibroblasts/drug effects , Humans , Neovascularization, Pathologic/drug therapy , Serum Albumin/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Statistics, Nonparametric
4.
Protein Pept Lett ; 16(11): 1414-23, 2009.
Article in English | MEDLINE | ID: mdl-19594432

ABSTRACT

The interaction of calreticulin with native and denatured forms and polypeptides in proteolytic digests of proteins representing structural classes of all-alpha-helix (hemoglobin, serum albumin), all-beta-sheet (IgG) and alpha-helix + beta-sheets (lysozyme, ovalbumin) was investigated. The binding of calreticulin to denatured proteins was found to depend on conformation and structural class of the protein. No interaction was observed with the native proteins, whereas binding was seen for the denatured proteins, the order of interaction being lysozyme = IgG > ovalbumin >> hemoglobin = serum albumin. Moreover, the interaction between calreticulin and the heat-denatured proteins depended on the temperature and time used for denaturation and the degree of proteolytic fragmentation. Calreticulin bound well to peptides in proteolytic digests from protease K or chymotrypsin treatment of lysozyme, IgG and ovalbumin but weakly or not at all to peptides in proteolytic digests of hemoglobin and serum albumin. Synthetic peptides from lysozyme and ovalbumin confirmed binding to hydrophobic peptides from these proteins. These results show that calreticulin has the ability to interact with denatured and fragmented forms of proteins with a preference for beta-strand structure and hydrophobicity.


Subject(s)
Calreticulin/metabolism , Peptides/metabolism , Proteins/metabolism , Calreticulin/chemistry , Hemoglobins/chemistry , Hemoglobins/metabolism , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Muramidase/chemistry , Muramidase/metabolism , Ovalbumin/chemistry , Ovalbumin/metabolism , Peptides/chemistry , Pregnancy Proteins/chemistry , Pregnancy Proteins/metabolism , Protein Binding , Protein Denaturation , Protein Folding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proteins/chemistry , Serum Albumin/chemistry , Serum Albumin/metabolism , Temperature
5.
Scand J Clin Lab Invest ; 68(7): 548-54, 2008.
Article in English | MEDLINE | ID: mdl-18609089

ABSTRACT

OBJECTIVE: Tissue inhibitor of metalloproteinases 1 (TIMP-1) has been identified as a potential biomarker in diseases such as cancer, cardiovascular diseases and diabetes. Since TIMP-1 resides in most tissues and bodily fluids, we evaluated the potential of using saliva to obtain reproducible TIMP-1 measurements in a non-invasive manner. MATERIAL AND METHODS: Samples of unstimulated and stimulated whole saliva and saliva collected from individual glands were analysed for TIMP-1 content. A TIMP-1 ELISA was validated for use in saliva testing and the most optimal sampling and handling procedures for reproducible measurements identified. Western blotting and MALDI-TOF mass spectrometry were used for confirmatory analyses. RESULTS: The TIMP-1 ELISA was found suitable for saliva measurements. All saliva secretions contained TIMP-1, but in different concentrations ranging from 2.81 ng/mL in submandibular/sublingual saliva to 173.88 ng/mL in parotid saliva. TIMP-1 concentrations were influenced to a varying degree by fluctuations in flow. We found the lowest output in submandibular/sublingual saliva stimulated with 0.5% citric acid (3.56 ng/min) and highest output in chewing-stimulated whole saliva (267.01 ng/min). CONCLUSION: This study shows that saliva contains authentic TIMP-1, the concentration of which was found to depend on gland type and salivary flow. Stimulated whole saliva is suggested as a reliable and easily accessible source for TIMP-1 determinations in bodily fluids.


Subject(s)
Parotid Gland/metabolism , Saliva/chemistry , Tissue Inhibitor of Metalloproteinase-1/metabolism , Female , Humans , Male , Mastication , Parotid Gland/chemistry , Salivary Glands, Minor/metabolism
6.
Scand J Immunol ; 66(5): 501-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17953526

ABSTRACT

The molecular chaperone calreticulin has been shown to bind C1q and mannan-binding lectin (MBL), which are constituents of the innate immune defence system. C1q and MBL do not share a large sequence identity but have a similar overall molecular architecture: an N-terminal triple-helical collagen-like domain and a C-terminal globular domain with ligand-binding properties. C1q is a hetero-trimer, while MBL is a homo-trimer, but due to the presence of N-terminal cysteines they both form higher order oligomers of trimers, which are the mature functional molecules. The same molecular architecture is utilized by many other functionally diverse molecules and in this work the interaction of calreticulin with C1q and structurally similar molecules was investigated. In addition to C1q and MBL, CD40 ligand (CD40L), tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) were found to bind calreticulin strongly. A low level or no binding was observed for adiponectin, tumour necrosis factor-alpha (TNF-alpha), CD30L, surfactant protein-A and -D and collagen VIII. The interaction with calreticulin required a conformational change in CD40L, TRAIL and FasL and showed the same characteristics as calreticulin's interaction with C1q and MBL: a time-dependent saturable binding to immobilized protein, which was initially sensitive to salt but gradually developed into a salt-insensitive interaction. Thus, the interaction requires a structural change in the interaction partner and leads to a conformational change in calreticulin itself. The implications of these results are that calreticulin may function as a general response modifier for a whole group of immunologically important proteins.


Subject(s)
CD40 Ligand/metabolism , Calreticulin/metabolism , Fas Ligand Protein/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adiponectin/chemistry , Adiponectin/immunology , Adiponectin/metabolism , CD40 Ligand/chemistry , CD40 Ligand/immunology , Calreticulin/chemistry , Calreticulin/immunology , Complement C1q/chemistry , Complement C1q/immunology , Complement C1q/metabolism , Fas Ligand Protein/chemistry , Fas Ligand Protein/immunology , Humans , Surface Plasmon Resonance , TNF-Related Apoptosis-Inducing Ligand/chemistry , TNF-Related Apoptosis-Inducing Ligand/immunology , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
7.
Vox Sang ; 92(4): 338-50, 2007 May.
Article in English | MEDLINE | ID: mdl-17456158

ABSTRACT

BACKGROUND AND OBJECTIVES: Mannan-binding lectin (MBL) is an important component of the innate immune defence; it binds to carbohydrate structures on pathogenic micro-organisms resulting in complement activation and opsonization. Individuals with low MBL levels are at risk of recurrent and severe infections. Substitution therapy with plasma-derived MBL is a promising treatment of diseases associated with MBL deficiency. A first-generation MBL product has been shown to be safe and well tolerated, and patients have benefited from MBL treatment. Following is a description of the development of a nanofiltered second-generation MBL product from Cohn fraction III, with the use of a new affinity matrix for MBL purification and the characteristics of this improved product. MATERIALS AND METHODS: Carbohydrate-based gels were comparatively screened as affinity matrices. MBL was extracted from fraction III, and affinity purified on a Superdex 200 pg column. The eluted material underwent two virus reduction steps: filtration through Planova 20N and solvent/detergent treatment. It was further purified by anion-exchange and gel-filtration chromatography. The affinity eluate and the final MBL fraction were characterized by protein chemical, immunological, and functional assays. RESULTS: In production scale, Superdex 200 pg was found to be superior to other carbohydrate-based matrices, and MBL was affinity purified from fraction III with a yield of 70%. The viral safety was increased by performing a nanofiltration of the affinity eluate through Planova 20N with a minimal loss of MBL. The purity of the final MBL fraction was 53% excluding the MBL-associated serine proteases (MASP). The product consisted of high-oligomeric MBL, with two dominating forms, and with MASP-1, -2, -3 and 19 kDa MBL-associated protein (MAp19). Only a few protein impurities were present, the major being alpha2-macroglobulin. MBL formed complexes with alpha2-macroglobulin bridged by MASP-1 covalently attached to the latter. The functional activity, assessed by mannan-binding activity and opsonic function, was intact, whereas half of the C4 activating capacity was lost during the production process. CONCLUSION: A second-generation MBL process was developed with an average yield of 50%. It was possible to nanofilter the MBL-MASP complexes through Planova 20N with only a minor loss resulting in an increased safety profile of this MBL product.


Subject(s)
Mannose-Binding Lectin/isolation & purification , Amino Acid Sequence , Animals , Antibodies , Chromatography, Affinity/methods , Filtration/methods , Humans , Immunity, Innate , In Vitro Techniques , Mannose-Binding Lectin/blood , Mannose-Binding Lectin/therapeutic use , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Molecular Sequence Data , Nanotechnology , Plasma/chemistry , Plasma/immunology , Rabbits , Safety , Viruses/isolation & purification
8.
Int J Biochem Cell Biol ; 33(6): 577-89, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11378439

ABSTRACT

We previously identified the human Fas associated factor (FAF1) as one of the interacting partners of protein kinase CK2 beta subunit. Since FAF1 is a phosphoprotein we investigated whether it is a substrate for CK2. Here, we report the full length human FAF1 cDNA sequence, expression of FAF1 in Escherichia coli and purification and characterization of FAF1 as a substrate for CK2. FAF1 as well as an N-terminal 40 kDa degradation product serve as substrates for both the recombinant CK2 holoenzyme (km 100 microM) and the isolated catalytic alpha subunit (km 200 microM). Despite the high k(m) values, we obtained evidence that CK2 is the major cellular kinase responsible for FAF1 phosphorylation, using tissue extracts as kinase sources. By MALDI-MS we identified the two serine residues at positions 289 and 291 as the major in vitro CK2 phosphorylation sites. These data may help us elucidate the functions of FAF1 and the involvement of CK2 mediated phosphorylation in processes such as apoptotic signaling, ubiquitination, nuclear translocation and embryonic development.


Subject(s)
Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Base Sequence , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Casein Kinase II , DNA, Complementary , Gene Expression , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Serine/metabolism , Tissue Extracts
9.
Eur J Biochem ; 268(9): 2558-65, 2001 May.
Article in English | MEDLINE | ID: mdl-11322874

ABSTRACT

The domain organization and the post-translational modifications of human placenta calreticulin were analysed by MS in combination with proteolytic digestion. Prolonged treatment with trypsin, chymotrypsin, elastase, Staphylococcus aureus V8 protease, or proteinase K all led to a 6- to 7-kDa decrease in the molecular mass of calreticulin. The decrease was found to be due to cleavages in the region around residue 340. In addition, minor fragments resulting from secondary cleavages close to the N-terminus were observed, but no stable fragments of intermediate size were found. These results show that the C-domain of calreticulin is susceptible to proteolytic cleavage and that the N- and P-domains form a proteolytically stable tight association. A disulfide bridge between the first two cysteines was mapped in the N-domain, and the third cysteine was found in the reduced form. No post-translational modifications in the form of glycosylation or phosphorylation were found. A modified form of calreticulin lacking the C-terminal hexapeptide including the KDEL endoplasmic reticulum retention sequon was isolated. Such a truncation may point to a mechanism that allows escape of calreticulin from the endoplasmic reticulum.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Placenta/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Amino Acid Sequence , Calreticulin , Endoplasmic Reticulum/metabolism , Female , Humans , In Vitro Techniques , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Pregnancy , Protein Processing, Post-Translational , Protein Structure, Tertiary , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Peptides ; 21(10): 1519-26, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11068099

ABSTRACT

Human insulin was glycated under hyperglycemic reducing conditions and a novel diglycated form (M(r) 6135.1 Da) was purified by RP-HPLC. Endoproteinase Glu-C digestion combined with mass spectrometry and automated Edman degradation localized glycation to Gly(1) and Phe(1) of the insulin A- and B-chains, respectively. Intraperitoneal (i.p.) administration of diglycated insulin to mice alone or in combination with glucose (7 nmol/kg) resulted in a 43-61% and 11-34% reduction in glucose lowering activity, respectively, compared with native insulin. Consistent with these findings, diglycated insulin (10(-9) to 10(-7) mol/liter) was 22-38% less effective (P < 0.001) than native insulin in stimulating glucose uptake, glucose oxidation and glycogen production in isolated mouse abdominal muscle.


Subject(s)
Hyperglycemia/drug therapy , Insulin/analogs & derivatives , Insulin/pharmacology , Amino Acids/analysis , Animals , Biological Transport/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Chromatography, High Pressure Liquid , Deoxyglucose/metabolism , Glucose/metabolism , Glucose/pharmacology , Glycogen/biosynthesis , Glycoproteins/chemistry , Glycoproteins/isolation & purification , Glycoproteins/pharmacology , Glycoproteins/therapeutic use , Glycosylation , Humans , Hyperglycemia/blood , Hyperglycemia/metabolism , Injections, Intraperitoneal , Insulin/chemistry , Insulin/therapeutic use , Male , Mass Spectrometry , Mice , Mice, Inbred Strains , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Serine Endopeptidases/metabolism
11.
Biochim Biophys Acta ; 1543(1): 159-73, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11087951

ABSTRACT

Human surfactant protein-A (SP-A) has been purified from a proteinosis patient and characterised by a combination of automated Edman degradation and mass spectrometry. The complete protein sequence was characterised. The major part of SP-A was shown to consist of SP-A2 gene product, and only a small amount of SP-A1 gene product was shown to be present. A cysteine extension to the N-terminal was indicated by sequence data, but was not definitely proven. All proline residues in the Y position of Gly-X-Y in the collagen-like region were at least partially modified to hydroxy-proline, but no lysine residues were found to be modified. A complex N-linked glycosylation was found on Asn-187 showing great heterogeneity as variants from a mono-antennary to penta-antennary glycosylation with varying amounts of attached pentose were identified. The disulfide bridges in the carbohydrate recognition domain were identified to be in the 1-4, 2-3 pattern common for collectins. Interchain disulfide bridges were discovered between two Cys-48 residues and cysteine residues in the N-terminal region. However, the exact disulfide bridge connections within the bouquet-like ultrastructure could not be established.


Subject(s)
Proteolipids/chemistry , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Surfactants/chemistry , Amino Acid Sequence , Bronchoalveolar Lavage Fluid/chemistry , Chromatography, High Pressure Liquid , Disulfides/chemistry , Humans , Molecular Sequence Data , Molecular Weight , Polysaccharides/chemistry , Proteolipids/isolation & purification , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Proteins , Pulmonary Surfactants/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin
12.
Eur J Biochem ; 267(10): 2945-54, 2000 May.
Article in English | MEDLINE | ID: mdl-10806393

ABSTRACT

Calreticulin is a highly conserved eukaryotic ubiquitious protein located mainly in the endoplasmic reticulum. Two major characteristics of calreticulin are its chaperone activity and its lectin properties, but its precise function in intracellular protein and peptide processing remains to be elucidated. We have investigated the interactions of human calreticulin with denatured ovalbumin, proteolytic digests of ovalbumin, and different available peptides by solid phase assays, size-exclusion chromatography, capillary electrophoresis, and MS. The results show that calreticulin interacts better with unfolded ovalbumin than with native ovalbumin, that calreticulin strongly binds components in proteolytic digests of denatured ovalbumin, and that calreticulin interacts strongly with certain synthetic peptides.


Subject(s)
Calcium-Binding Proteins/metabolism , Molecular Chaperones/metabolism , Peptides/metabolism , Ribonucleoproteins/metabolism , Amino Acid Sequence , Biotinylation , Calreticulin , Chromatography, Gel , Electrophoresis, Capillary , Electrophoresis, Polyacrylamide Gel , Endopeptidase K/metabolism , Enzyme-Linked Immunosorbent Assay , Hot Temperature , Humans , Molecular Sequence Data , Ovalbumin/metabolism , Placenta/chemistry , Protein Binding , Protein Denaturation , Protein Folding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors
13.
Anal Biochem ; 274(2): 153-62, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10527511

ABSTRACT

A new method combining protein chemistry with enzymes immobilized to paramagnetic beads is presented. The immobilized enzymes can substitute for regular enzymes in a number of protein chemistry protocols, resulting in faster reaction times, less sample contamination, and improved interfacing to modern procedures, like mass spectrometry. Trypsin was used as a model enzyme to test the amount of protein coupled to glass beads and the degree of autodigestion when analyzed by MALDI-MS and HPLC. Immobilization of trypsin resulted in digestions comparable with standard solution digestions using fetuin as a model substrate. Furthermore, fetuin was used to test the stability of the enzyme-coated beads. No apparent loss of enzyme activity was observed after 10 times reuse of trypsin-coated beads. Immobilization of exo- and endoglycosidases to paramagnetic beads resulted in high sensitivity, faster sequential glycosidase digestion of glycopeptides, and reduced sample contamination. All digestions could be performed in less than 24 h, when a tryptic glycopeptide from human lung proteinosis surfactant protein A was used as model compound.


Subject(s)
Chromatography, Affinity/methods , Enzymes, Immobilized , Proteins/analysis , Animals , Chromatography, High Pressure Liquid , Humans , Magnetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Swine
14.
Infect Immun ; 67(11): 5552-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531199

ABSTRACT

Mycobacterium tuberculosis GroES was purified from culture filtrate, and its identity was confirmed by immunoblot analysis and N-terminal sequencing. Comparing the immunological recognition of native and recombinant GroES, we found that whereas native GroES elicited a strong proliferative response and release of gamma interferon-gamma by peripheral blood mononuclear cells from healthy tuberculin reactors, the recombinant protein failed to do so. The same difference in immunological recognition was observed in a mouse model of TB infection. Both the native and recombinant preparations were recognized by mice immunized with the recombinant protein. Biochemical characterization including sodium dodecyl sulfate-polyacrylamide gel electrophoresis, two-dimensional electrophoresis, and mass spectrometry analysis of both proteins demonstrated no differences between the native and recombinant forms of GroES except for the eight additional N-terminal amino acids derived from the fusion partner in recombinant GroES. The recombinant fusion protein, still tagged with the maltose binding protein, was recognized by T cells isolated from TB-infected mice if mixed with culture filtrate before affinity purification on an amylose column. The maltose binding protein treated in the same manner as a control preparation was not recognized. Based on the data presented, we suggest that the association of biologically active molecules from culture filtrate with the chaperone GroES may be responsible for the observed T-cell recognition of the native preparation.


Subject(s)
Chaperonin 10/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Animals , Chaperonin 10/chemistry , Chaperonin 10/isolation & purification , Escherichia coli/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/immunology
15.
Biochem J ; 343 Pt 3: 645-52, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10527944

ABSTRACT

Human and bovine surfactant proteins D (SP-D) were purified from late amniotic fluid and bronchioalveolar lavage on the basis of its Ca(2+)-dependent affinity for maltose. The molecular mass of a trimeric subunit was determined by matrix-assisted laser desorption ionization MS to lie in the range 115-125 kDa for human SP-D and 110-123 kDa for bovine SP-D. A single polypeptide chain was determined at 37-41 and 36-40 kDa for the human and bovine species respectively. The major parts of the primary structures of both SP-D molecules were determined by a combination of MS and Edman degradation. The heterogeneity in SP-D was caused mainly by a high number of post-translational modifications in the collagen-like region. Proline and lysine residues were partly hydroxylated and lysine residues were further O-glycosylated with the disaccharide galactose-glucose. A partly occupied N-linked glycosylation site was characterized in human SP-D. The carbohydrate was determined as a complex type bi-antennary structure, with a small content of mono-antennary and tri-antennary structures. No sialic acid residues were present on the glycan, but some had an attached fucose and/or an N-acetylglucosamine residue linked to the core. Bovine SP-D was determined as having a similar structure.


Subject(s)
Amniotic Fluid/chemistry , Bronchoalveolar Lavage Fluid/chemistry , Glycoproteins/chemistry , Pulmonary Surfactants/chemistry , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cattle , Chromatography, High Pressure Liquid , Collagenases , Electrophoresis, Polyacrylamide Gel , Female , Glycoproteins/isolation & purification , Glycosylation , Humans , Molecular Sequence Data , Molecular Weight , Oxidation-Reduction , Peptide Fragments/chemistry , Pregnancy , Pulmonary Surfactant-Associated Protein D , Pulmonary Surfactants/isolation & purification , Sequence Alignment , Sequence Homology, Amino Acid , Trypsin
16.
J Biol Chem ; 274(36): 25481-9, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10464279

ABSTRACT

alpha-Synuclein has been implicated in the pathogenesis of several neurodegenerative disorders based on the direct linking of missense mutations in alpha-synuclein to autosomal dominant Parkinson's disease and its presence in Lewy-like lesions. To gain insight into alpha-synuclein functions, we have investigated whether it binds neuronal proteins and modulates their functional state. The microtubule-associated protein tau was identified as a ligand by alpha-synuclein affinity chromatography of human brain cytosol. Direct binding assays using (125)I-labeled human tau40 demonstrated a reversible binding with a IC(50) about 50 pM. The interacting domains were localized to the C terminus of alpha-synuclein and the microtubule binding region of tau as determined by protein fragmentation and the use of recombinant peptides. High concentrations of tubulin inhibited the binding between tau and alpha-synuclein. Functionally, alpha-synuclein stimulated the protein kinase A-catalyzed phosphorylation of tau serine residues 262 and 356 as determined using a phospho-epitope-specific antibody. We propose that alpha-synuclein modulates the phosphorylation of soluble axonal tau and thereby indirectly affects the stability of axonal microtubules.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , tau Proteins/metabolism , Axons/metabolism , Axons/pathology , Escherichia coli , Humans , Neurons/pathology , Phosphoproteins/metabolism , Phosphorylation , Serine , Signal Transduction , Synucleins , alpha-Synuclein
17.
J Biol Chem ; 274(24): 16953-8, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10358043

ABSTRACT

The covalent heme attachment has been extensively studied by spectroscopic methods in myeloperoxidase and lactoperoxidase (LPO) but not in eosinophil peroxidase (EPO). We show that heme linkage to the heavy chain is invariably present, whereas heme linkage to the light chain of EPO is present in less than one-third of EPO molecules. Mass analysis of isolated heme bispeptides supports the hypothesis of a heme b linked through two esters to the polypeptide. Mass analysis of heme monopeptides reveals that >90% have a nonderivatized methyl group at the position of the light chain linkage. Apparently, an ester had not been formed during biosynthesis. The light chain linkage could be formed by incubation with hydrogen peroxide, in accordance with a recent hypothesis of autocatalytic heme attachment based on studies with LPO (DePillis, G. D., Ozaki, S., Kuo, J. M., Maltby, D. A., and Ortiz de Montellano P. R. (1997) J. Biol. Chem. 272, 8857-8860). By sequence analysis of isolated heme peptides after aminolysis, we unambiguously identified the acidic residues, Asp-93 of the light chain and Glu-241 of the heavy chain, that form esters with the heme group. This is the first biochemical support for ester linkage to two specific residues in eosinophil peroxidase. From a parallel study with LPO, we show that Asp-125 and Glu-275 are engaged in ester linkage. The species with a nonderivatized methyl group was not found among LPO peptides.


Subject(s)
Aspartic Acid/chemistry , Eosinophils/enzymology , Glutamic Acid/chemistry , Heme/chemistry , Peroxidases/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Eosinophil Peroxidase , Esters , Humans , Lactoperoxidase/chemistry , Molecular Sequence Data , Oligopeptides/chemistry , Sequence Analysis
18.
J Biol Chem ; 274(21): 14818-22, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10329680

ABSTRACT

Extracellular superoxide dismutase (EC-SOD) is the only known extracellular enzyme designed to scavenge the superoxide anion. The purified enzyme exists in two forms when visualized by reduced SDS-polyacrylamide gel electrophoresis: (i) intact EC-SOD (Trp1-Ala222) containing the C-terminal heparin-binding domain and (ii) cleaved EC-SOD (Trp1-Glu209) without the C-terminal heparin-binding domain. The proteolytic event(s) leading to proteolysis at Glu209-Arg210 and removal of the heparin-binding domain are not known, but may represent an important regulatory mechanism. Removal of the heparin-binding domain affects both the affinity of EC-SOD for and its distribution to the extracellular matrix, in which it is secreted. During the purification of human EC-SOD, the intact/cleaved ratio remains constant, suggesting that proteolytic removal of the heparin-binding domain does not occur during purification (Oury, T. D., Crapo, J. D., Valnickova, Z., and Enghild, J. J. (1996) Biochem. J. 317, 51-57). This was supported by the finding that fresh mouse tissue contains both intact and cleaved EC-SOD. To study other possible mechanisms leading to the formation of cleaved EC-SOD, we examined biosynthesis in cultured rat L2 epithelial-like cells using a pulse-chase protocol. The results of these studies suggest that the heparin-binding domain is removed intracellularly just prior to secretion. In addition, the intact/cleaved EC-SOD ratio appears to be tissue-dependent, implying that the intracellular processing event is regulated in a tissue-specific manner. The existence of this intracellular processing pathway may thus represent a novel regulatory pathway for affecting the distribution and effect of EC-SOD.


Subject(s)
Heparin/pharmacokinetics , Protein Splicing , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/metabolism , Animals , Culture Techniques , Extracellular Space/metabolism , Humans , Mice , Rats
19.
Proc Natl Acad Sci U S A ; 96(6): 2811-6, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077593

ABSTRACT

Angiostatin, a proteolytic fragment of plasminogen, is a potent antagonist of angiogenesis and an inhibitor of endothelial cell migration and proliferation. To determine whether the mechanism by which angiostatin inhibits endothelial cell migration and/or proliferation involves binding to cell surface plasminogen receptors, we isolated the binding proteins for plasminogen and angiostatin from human umbilical vein endothelial cells. Binding studies demonstrated that plasminogen and angiostatin bound in a concentration-dependent, saturable manner. Plasminogen binding was unaffected by a 100-fold molar excess of angiostatin, indicating the presence of a distinct angiostatin binding site. This finding was confirmed by ligand blot analysis of isolated human umbilical vein endothelial cell plasma membrane fractions, which demonstrated that plasminogen bound to a 44-kDa protein, whereas angiostatin bound to a 55-kDa species. Amino-terminal sequencing coupled with peptide mass fingerprinting and immunologic analyses identified the plasminogen binding protein as annexin II and the angiostatin binding protein as the alpha/beta-subunits of ATP synthase. The presence of this protein on the cell surface was confirmed by flow cytometry and immunofluorescence analysis. Angiostatin also bound to the recombinant alpha-subunit of human ATP synthase, and this binding was not inhibited by a 2,500-fold molar excess of plasminogen. Angiostatin's antiproliferative effect on endothelial cells was inhibited by as much as 90% in the presence of anti-alpha-subunit ATP synthase antibody. Binding of angiostatin to the alpha/beta-subunits of ATP synthase on the cell surface may mediate its antiangiogenic effects and the down-regulation of endothelial cell proliferation and migration.


Subject(s)
Adenosine Triphosphatases/metabolism , Antineoplastic Agents/metabolism , Endothelium, Vascular/metabolism , Peptide Fragments/metabolism , Plasminogen/metabolism , Angiostatins , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Membrane Proteins/metabolism , Neovascularization, Pathologic/prevention & control , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Protein Binding
20.
Anal Chem ; 71(5): 919-27, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10079758

ABSTRACT

A method for mass spectrometric peptide mapping was developed, based on hydrolysis of a solid protein by acid vapor followed by mass spectrometric analysis of the cleavage products. The method is applicable to lyophilized samples as well as proteins present in gels after separation by SDS-PAGE. The cleavage specificity was established using a number of standard proteins. Three different types of cleavages were observed: specific internal backbone cleavages at Asp, Ser, Thr, and Gly and N- and C-terminal sequence ladders. On the basis of the observed cleavage characteristics, a strategy for protein identification based on the peptide mass maps was developed. The identification strategy utilizes the specific internal backbone cleavages as well as the partial sequence information, obtained from the sequence ladders.


Subject(s)
Peptide Mapping/methods , Proteins/analysis , Amino Acid Sequence , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Molecular Sequence Data , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...