Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(7): e0265554, 2022.
Article in English | MEDLINE | ID: mdl-35839162

ABSTRACT

BACKGROUND: Pulmonary Tuberculosis (TB) is diagnosed through sputum samples. As sputum sampling is challenging in children and cachexic patients, the development of diagnostic tests using saliva appears promising but has been discouraged due to low bacterial load and poor sensitivity. Here, we present a novel and rapid method to enrich Mycobacterium tuberculosis (Mtb) from saliva, which may serve as a basis for a diagnostic saliva test. METHODS: Lipobiotin-functionalized magnetic beads (LMBs) were incubated with Mtb-spiked PBS and saliva from healthy donors as well as with saliva from TB patients. Flow cytometry was used to evaluate the capacity of the beads to bind Mtb, while real-time quantitative polymerase chain reaction (qPCR) was utilized to detect Mtb and determine the amount of mycobacterial DNA in different sample types. RESULTS: We found that LMBs bind Mtb efficiently when compared to non-functionalized beads. The development of an qPCR assay based on the use of LMBs (LMB assay) allowed us to enrich mycobacterial DNA in spiked sample types, including PBS and saliva from healthy donors (enrichment of up to ~8.7 fold). In Mtb-spiked saliva samples, we found that the LMB assay improved the detection rate of 102 bacteria in a volume of 5 ml from 0 out of 15 (0%) to 6 out of 15 (40%). Consistent with that, the LMB assay increased the rate of correctly identified saliva samples from TB patients in two independent cohorts. CONCLUSIONS: Implementation of the principle of the LMB-based assay may improve the sensitivity of existing diagnostic techniques, e.g. by functionalizing materials that facilitate Mtb sampling from the oral cavity.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis, Lymph Node , Tuberculosis, Pulmonary , Child , Humans , Magnetic Phenomena , Mycobacterium tuberculosis/genetics , Saliva , Sensitivity and Specificity , Sputum/microbiology , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/microbiology
2.
J Biol Chem ; 298(5): 101901, 2022 05.
Article in English | MEDLINE | ID: mdl-35395245

ABSTRACT

Conformational flexibility in antibody-combining sites has been hypothesized to facilitate polyspecificity toward multiple unique epitopes and enable the limited germline repertoire to match an overwhelming diversity of potential antigens; however, elucidating the mechanisms of antigen recognition by flexible antibodies has been understandably challenging. Here, multiple liganded and unliganded crystal structures of the near-germline anticarbohydrate antibodies S25-2 and S25-39 are reported, which reveal an unprecedented diversity of complementarity-determining region H3 conformations in apparent equilibrium. These structures demonstrate that at least some germline or near-germline antibodies are flexible entities sensitive to their chemical environments, with conformational selection available as an evolved mechanism that preserves the inherited ability to recognize common pathogens while remaining adaptable to new threats.


Subject(s)
Antibodies , Complementarity Determining Regions , Antibodies/chemistry , Binding Sites, Antibody , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Crystallography, X-Ray , Germ Cells , Molecular Conformation , Protein Conformation
3.
Sci Adv ; 5(2): eaav2554, 2019 02.
Article in English | MEDLINE | ID: mdl-30788437

ABSTRACT

Influenza A viruses can bind sialic acid-terminating glycan receptors, and species specificity is often correlated with sialic acid linkage with avian strains recognizing α2,3-linked sialylated glycans and mammalian strains preferring α2,6-linked sialylated glycans. These paradigms derive primarily from studies involving erythrocyte agglutination, binding to synthetic receptor analogs or binding to undefined surface markers on cells or tissues. Here, we present the first examination of the N-glycome of the human lung for identifying natural receptors for a range of avian and mammalian influenza viruses. We found that the human lung contains many α2,3- and α2,6-linked sialylated glycan determinants bound by virus, but all viruses also bound to phosphorylated, nonsialylated glycans.


Subject(s)
Influenza A virus/physiology , Influenza, Human/metabolism , Influenza, Human/virology , Lung/metabolism , Lung/virology , Polysaccharides/metabolism , Animals , Chromatography, High Pressure Liquid , Humans , Mass Spectrometry , Phosphorylation , Polysaccharides/chemistry , Proteomics/methods , Viral Proteins
4.
Biochemistry ; 58(6): 714-726, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30571096

ABSTRACT

Murine antibodies S25-23, S25-26, and S25-5 derive from a common germ-line origin, and all bind the Chlamydiaceae family-specific epitope αKdo(2→8)αKdo(2→4)αKdo (where Kdo is 3-deoxy-α-d- manno-oct-2-ulosonic acid) with high affinity and specificity. These antibodies recognize the entire trisaccharide antigen in a linkage-dependent manner via a groove composed largely of germ-line residues. Despite sharing identical heavy and light chain genes, S25-23 binds the family-specific epitope with nanomolar affinity, which is an order of magnitude higher than that of S25-26, while S25-5 displays an affinity between those of S25-23 and S25-26. We determined the high-resolution crystal structures of S25-23 and S25-5 antigen binding fragments in complex with a pentasaccharide derived from the LPS of Chlamydia and measured the affinity of S25-5 for chlamydial LPS antigens using isothermal titration microcalorimetry. The 1.75 Å resolution structure of S25-23 shows how subtle conservative mutations Arg(L)-27E to lysine and Ser(H)-56 to threonine lead to an order of magnitude increase in affinity. Importantly, comparison between previous S25-26 structures and the 1.99 and 2.05 Å resolution liganded and unliganded structures of S25-5, respectively, shows how a Ser(L)-27E mutation results in an intermediate affinity due to the reduced enthalpic penalty associated with complex formation that would otherwise be required for arginine in this position. This strategy allows for subtle adjustments in the combining site via affinity maturation that have dramatic consequences for the affinity of an antibody for its antigen.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/metabolism , Chlamydiaceae/immunology , Epitopes/metabolism , Oligosaccharides/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Monoclonal, Murine-Derived/immunology , Antibody Affinity , Binding Sites, Antibody , Epitopes/immunology , Hydrogen Bonding , Mice , Oligosaccharides/immunology , Protein Binding , Sequence Alignment
5.
Mol Cell Proteomics ; 17(8): 1612-1626, 2018 08.
Article in English | MEDLINE | ID: mdl-29773673

ABSTRACT

Targeting of soluble lysosomal enzymes requires mannose 6-phosphate (M6P) signals whose formation is initiated by the hexameric N-acetylglucosamine (GlcNAc)-1-phosphotransferase complex (α2ß2γ2). Upon proteolytic cleavage by site-1 protease, the α/ß-subunit precursor is catalytically activated but the functions of γ-subunits (Gnptg) in M6P modification of lysosomal enzymes are unknown. To investigate this, we analyzed the Gnptg expression in mouse tissues, primary cultured cells, and in Gnptg reporter mice in vivo, and found high amounts in the brain, eye, kidney, femur, vertebra and fibroblasts. Consecutively we performed comprehensive quantitative lysosomal proteome and M6P secretome analysis in fibroblasts of wild-type and Gnptgko mice mimicking the lysosomal storage disorder mucolipidosis III. Although the cleavage of the α/ß-precursor was not affected by Gnptg deficiency, the GlcNAc-1-phosphotransferase activity was significantly reduced. We purified lysosomes and identified 29 soluble lysosomal proteins by SILAC-based mass spectrometry exhibiting differential abundance in Gnptgko fibroblasts which was confirmed by Western blotting and enzymatic activity analysis for selected proteins. A subset of these lysosomal enzymes show also reduced M6P modifications, fail to reach lysosomes and are secreted, among them α-l-fucosidase and arylsulfatase B. Low levels of these enzymes correlate with the accumulation of non-degraded fucose-containing glycostructures and sulfated glycosaminoglycans in Gnptgko lysosomes. Incubation of Gnptgko fibroblasts with arylsulfatase B partially rescued glycosaminoglycan storage. Combinatorial treatments with other here identified missorted enzymes of this degradation pathway might further correct glycosaminoglycan accumulation and will provide a useful basis to reveal mechanisms of selective, Gnptg-dependent formation of M6P residues on lysosomal proteins.


Subject(s)
Enzymes/metabolism , Lysosomes/metabolism , Mucolipidoses/metabolism , Mucolipidoses/pathology , Proteome/metabolism , Animals , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Glycosaminoglycans/metabolism , Humans , Isotope Labeling , Mannosephosphates/metabolism , Mice, Knockout , Protein Subunits/metabolism , Proteolysis , Substrate Specificity
6.
Adv Exp Med Biol ; 966: 181-202, 2017.
Article in English | MEDLINE | ID: mdl-28887790

ABSTRACT

The process of natural selection favours germ-line gene segments that encode CDRs that have the ability to recognize a range of structurally related antigens. This presents an immunological advantage to the host, as it can confer protection against a common pathogen and still cope with new or changing antigens. Cross-reactive and polyspecific antibodies also play a central role in autoimmune responses, and a link has been shown to exist between auto-reactive B cells and certain bacterial infections. Bacterial DNA, lipids, and carbohydrates have been implicated in the progression of autoimmune diseases such as systemic lupus erythematosus. As well, reports of anti-lipid A antibody polyspecificity towards single-stranded DNA together with the observed sequence homology amongst isolated auto- and anti-lipid A antibodies has prompted further study of this phenomenon. Though the lipid A epitope appears cryptic during Gram-negative bacterial infection, there have been several reported instances of lipid A-specific antibodies isolated from human sera, some of which have exhibited polyspecificity for single stranded DNA. In such cases, the breakdown of negative selection through polyspecificity can have the unfortunate consequence of autoimmune disease. This review summarizes current knowledge regarding such antibodies and emphasizes the features of S1-15, A6, and S55-5, anti-lipid A antibodies whose structures were recently determined by X-ray crystallography.


Subject(s)
Antibody Specificity , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmunity , Bacterial Infections/immunology , Lipid A/immunology , Animals , Autoantibodies/chemistry , Autoimmune Diseases/microbiology , B-Lymphocytes/immunology , Bacterial Infections/microbiology , DNA, Single-Stranded/immunology , Humans , Models, Molecular , Protein Conformation , Structure-Activity Relationship
7.
J Biol Chem ; 291(19): 10104-18, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26933033

ABSTRACT

Lipopolysaccharide dispersed in the blood by Gram-negative bacteria can be a potent inducer of septic shock. One research focus has been based on antibody sequestration of lipid A (the endotoxic principle of LPS); however, none have been successfully developed into a clinical treatment. Comparison of a panel of anti-lipid A antibodies reveals highly specific antibodies produced through distinct germ line precursors. The structures of antigen-binding fragments for two homologous mAbs specific for lipid A, S55-3 and S55-5, have been determined both in complex with lipid A disaccharide backbone and unliganded. These high resolution structures reveal a conserved positively charged pocket formed within the complementarity determining region H2 loops that binds the terminal phosphates of lipid A. Significantly, this motif occurs in unrelated antibodies where it mediates binding to negatively charged moieties through a range of epitopes, including phosphorylated peptides used in diagnostics and therapeutics. S55-3 and S55-5 have combining sites distinct from anti-lipid A antibodies previously described (as a result of their separate germ line origin), which are nevertheless complementary both in shape and charge to the antigen. S55-3 and S55-5 display similar avidity toward lipid A despite possessing a number of different amino acid residues in their combining sites. Binding of lipid A occurs independent of the acyl chains, although the GlcN-O6 attachment point for the core oligosaccharide is buried in the combining site, which explains their inability to recognize LPS. Despite their lack of therapeutic potential, the observed motif may have significant immunological implications as a tool for engineering recombinant antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Complementarity Determining Regions/immunology , Epitopes/immunology , Immunoglobulin Fab Fragments/immunology , Lipid A/immunology , Lipopolysaccharides/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/metabolism , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/metabolism , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Lipid A/chemistry , Lipid A/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Homology, Amino Acid
8.
Glycobiology ; 26(2): 181-92, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26503547

ABSTRACT

The acquisition of mannose 6-phosphate (Man6P) on N-linked glycans of lysosomal enzymes is a structural requirement for their transport from the Golgi apparatus to lysosomes mediated by the mannose 6-phosphate receptors, 300 kDa cation-independent mannose 6-phosphate receptor (MPR300) and 46 kDa cation-dependent mannose 6-phosphate receptor (MPR46). Here we report that the single-chain variable domain (scFv) M6P-1 is a unique antibody fragment with specificity for Man6P monosaccharide that, through an array-screening approach against a number of phosphorylated N-glycans, is shown to bind mono- and diphosphorylated Man6 and Man7 glycans that contain terminal αMan6P(1 → 2)αMan(1 → 3)αMan. In contrast to MPR300, scFv M6P-1 does not bind phosphodiesters, monophosphorylated Man8 or mono- or diphosphorylated Man9 structures. Single crystal X-ray diffraction analysis to 2.7 Å resolution of Fv M6P-1 in complex with Man6P reveals that specificity and affinity is achieved via multiple hydrogen bonds to the mannose ring and two salt bridges to the phosphate moiety. In common with both MPRs, loss of binding was observed for scFv M6P-1 at pH values below the second pKa of Man6P (pKa = 6.1). The structures of Fv M6P-1 and the MPRs suggest that the change of the ionization state of Man6P is the main driving force for the loss of binding at acidic lysosomal pH (e.g. lysosome pH ∼ 4.6), which provides justification for the evolution of a lysosomal enzyme transport pathway based on Man6P recognition.


Subject(s)
Mannosephosphates/chemistry , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Animals , Binding Sites , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Single-Chain Antibodies/metabolism
9.
J Biol Chem ; 290(32): 19629-40, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26085093

ABSTRACT

Septic shock is a leading cause of death, and it results from an inflammatory cascade triggered by the presence of microbial products in the blood. Certain LPS from Gram-negative bacteria are very potent inducers and are responsible for a high percentage of septic shock cases. Despite decades of research, mAbs specific for lipid A (the endotoxic principle of LPS) have not been successfully developed into a clinical treatment for sepsis. To understand the molecular basis for the observed inability to translate in vitro specificity for lipid A into clinical potential, the structures of antigen-binding fragments of mAbs S1-15 and A6 have been determined both in complex with lipid A carbohydrate backbone and in the unliganded form. The two antibodies have separate germ line origins that generate two markedly different combining-site pockets that are complementary both in shape and charge to the antigen. mAb A6 binds lipid A through both variable light and heavy chain residues, whereas S1-15 utilizes exclusively the variable heavy chain. Both antibodies bind lipid A such that the GlcN-O6 attachment point for the core oligosaccharide is buried in the combining site, which explains the lack of LPS recognition. Longstanding reports of polyspecificity of anti-lipid A antibodies toward single-stranded DNA combined with observed homology of S1-15 and A6 and the reports of several single-stranded DNA-specific mAbs prompted the determination of the structure of S1-15 in complex with single-stranded DNA fragments, which may provide clues about the genesis of autoimmune diseases such as systemic lupus erythematosus, thyroiditis, and rheumatic autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/chemistry , Glycoconjugates/chemistry , Immunoglobulin Fab Fragments/chemistry , Lipid A/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibody Specificity , Ascites/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Binding Sites, Antibody , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/immunology , Glycoconjugates/biosynthesis , Glycoconjugates/immunology , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/immunology , Lipid A/immunology , Mice , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Alignment , Sequence Homology, Amino Acid , Static Electricity
10.
Carbohydr Res ; 391: 66-81, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24785390

ABSTRACT

The α-d-glucopyranosyl-(1→5)-substituted methyl glycosides of 3-deoxy-α-d-manno-oct-2-ulosonic acid (Kdo), 3-deoxy-α-d-lyxo-hept-2-ulosonic acid (Kdh), and d-glycero-α-d-talo-oct-2-ulosonic acid (Ko) were prepared using orthogonally protected glycosyl acceptor derivatives via glycosylation with a torsionally disarmed 4,6-O-benzylidene protected trifluoroacetimidate glucosyl donor followed by global deprotection. The related 6-O-phosphoryl-α-d-glucopyranosyl-(1→5)-substituted Kdo and Kdh derivatives were derived from a benzylidene-protected glucosyl intermediate using phosphoramidite and phosphoryl chloride-based phosphorylation steps, respectively. The deprotected disaccharides serve as ligands to study lectin binding of Acinetobacter lipopolysaccharide core oligosaccharides.


Subject(s)
Acinetobacter/chemistry , Disaccharides/chemistry , Glycosides/chemistry , Glycosides/chemical synthesis , Lipopolysaccharides/chemistry , Sugar Acids/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Molecular Sequence Data
11.
J Biol Chem ; 289(24): 16644-61, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24682362

ABSTRACT

The structure of the antigen binding fragment of mAb S25-26, determined to 1.95 Å resolution in complex with the Chlamydiaceae family-specific trisaccharide antigen Kdo(2→8)Kdo(2→4)Kdo (Kdo = 3-deoxy-α-d-manno-oct-2-ulopyranosonic acid), displays a germ-line-coded paratope that differs significantly from previously characterized Chlamydiaceae-specific mAbs despite being raised against the identical immunogen. Unlike the terminal Kdo recognition pocket that promotes cross-reactivity in S25-2-type antibodies, S25-26 and the closely related S25-23 utilize a groove composed of germ-line residues to recognize the entire trisaccharide antigen and so confer strict specificity. Interest in S25-23 was sparked by its rare high µm affinity and strict specificity for the family-specific trisaccharide antigen; however, only the related antibody S25-26 proved amenable to crystallization. The structures of three unliganded forms of S25-26 have a labile complementary-determining region H3 adjacent to significant glycosylation of the variable heavy chain on asparagine 85 in Framework Region 3. Analysis of the glycan reveals a heterogeneous mixture with a common root structure that contains an unusually high number of terminal αGal-Gal moieties. One of the few reported structures of glycosylated mAbs containing these epitopes is the therapeutic antibody Cetuximab; however, unlike Cetuximab, one of the unliganded structures in S25-26 shows significant order in the glycan with appropriate electron density for nine residues. The elucidation of the three-dimensional structure of an αGal-containing N-linked glycan on a mAb variable heavy chain has potential clinical interest, as it has been implicated in allergic response in patients receiving therapeutic antibodies.


Subject(s)
Binding Sites, Antibody , Chlamydia/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Lipopolysaccharides/chemistry , Amino Acid Sequence , Antibodies, Bacterial/chemistry , Antibodies, Bacterial/immunology , Antibody Affinity , Chlamydia/chemistry , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Lipopolysaccharides/immunology , Molecular Docking Simulation , Molecular Sequence Data
12.
Biotechnol Appl Biochem ; 61(2): 184-92, 2014.
Article in English | MEDLINE | ID: mdl-24033810

ABSTRACT

Bioreactor process changes can have a profound effect on the yield and quality of biotechnology products. Mannose-6-phosphate (M6P) glycan content and the enzymatic catalytic kinetic parameters are critical quality attributes (CQAs) of many therapeutic enzymes used to treat lysosomal storage diseases (LSDs). Here, we have evaluated the effect of adding butyrate to bioreactor production cultures of human recombinant ß-glucuronidase produced from CHO-K1 cells, with an emphasis on CQAs. The ß-glucuronidase produced in parallel bioreactors was quantified by capillary electrophoresis, the catalytic kinetic parameters were measured using steady-state analysis, and mannose-6-phosphorylation status was assessed using an M6P-specific single-chain antibody fragment. Using this approach, we found that butyrate treatment increased ß-glucuronidase production up to approximately threefold without significantly affecting the catalytic properties of the enzyme. However, M6P content in ß-glucuronidase was inversely correlated with the increased enzyme production induced by butyrate treatment. This assessment demonstrated that although butyrate dramatically increased ß-glucuronidase production in bioreactors, it adversely impacted the mannose-6-phosphorylation of this LSD therapeutic enzyme. This strategy may have utility in evaluating manufacturing process changes to improve therapeutic enzyme yields and CQAs.


Subject(s)
Bioreactors , Butyrates/pharmacology , Glucuronidase/biosynthesis , Lysosomal Storage Diseases/enzymology , Animals , Butyrates/chemistry , CHO Cells , Cricetinae , Cricetulus , Glucuronidase/therapeutic use , Humans , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/pathology , Mannosephosphates/chemistry , Mannosephosphates/pharmacology , Phosphorylation , Polysaccharides/chemistry
13.
J Biol Chem ; 288(12): 8111-8127, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23372159

ABSTRACT

It is well established that lipopolysaccharide (LPS) often carries nonstoichiometric substitutions in lipid A and in the inner core. In this work, the molecular basis of inner core alterations and their physiological significance are addressed. A new inner core modification of LPS is described, which arises due to the addition of glucuronic acid on the third heptose with a concomitant loss of phosphate on the second heptose. This was shown by chemical and structural analyses. Furthermore, the gene whose product is responsible for the addition of this sugar was identified in all Escherichia coli core types and in Salmonella and was designated waaH. Its deduced amino acid sequence exhibits homology to glycosyltransferase family 2. The transcription of the waaH gene is positively regulated by the PhoB/R two-component system in a growth phase-dependent manner, which is coordinated with the transcription of the ugd gene explaining the genetic basis of this modification. Glucuronic acid modification was observed in E. coli B, K12, R2, and R4 core types and in Salmonella. We also show that the phosphoethanolamine (P-EtN) addition on heptose I in E. coli K12 requires the product of the ORF yijP, a new gene designated as eptC. Incorporation of P-EtN is also positively regulated by PhoB/R, although it can occur at a basal level without a requirement for any regulatory inducible systems. This P-EtN modification is essential for resistance to a variety of factors, which destabilize the outer membrane like the addition of SDS or challenge to sublethal concentrations of Zn(2+).


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Ethanolamines/metabolism , Glucuronic Acid/metabolism , Heptoses/metabolism , Lipopolysaccharides/metabolism , Membrane Proteins/genetics , Carbohydrate Conformation , Carbohydrate Sequence , Cell Membrane/metabolism , Cell Membrane/physiology , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , Gene Expression Regulation, Bacterial , Glycosyltransferases/genetics , Lipopolysaccharides/chemistry , Membrane Proteins/metabolism , Membrane Proteins/physiology , Molecular Sequence Annotation , Molecular Sequence Data , Transcription, Genetic
14.
Article in English | MEDLINE | ID: mdl-23295476

ABSTRACT

The near-germline antibody S25-2 exhibits a remarkable cross-reactivity for oligosaccharides containing the bacterial lipopolysaccharide carbohydrate 3-deoxy-D-manno-oct-2-ulosonic acid (Kdo). The recent synthesis of a variety of Kdo analogues permits a detailed structural analysis of the importance of specific interactions in antigen recognition by S25-2. The Kdo disaccharide analogue Kdo-(2→4)-5,6-dehydro-Kdo lacks a 5-OH group on the second Kdo residue and has been cocrystallized with S25-2. The structure reveals that the modification of the Kdo residue at position 5 results in a rearrangement of intramolecular hydrogen bonds in the antigen that allows it to assume a novel conformation in the antibody-combining site. The cross-reactive binding of S25-2 to this synthetic ligand highlights the adaptability of this antibody to non-natural synthetic analogues.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Sugar Acids/chemistry , Sugar Acids/immunology , Antibodies, Monoclonal/metabolism , Antigen-Antibody Reactions , Binding Sites, Antibody/immunology , Carbohydrate Conformation , Cross Reactions , Hydrogen Bonding , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Ligands , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Models, Molecular , Oligosaccharides/chemistry , Oligosaccharides/immunology , Protein Conformation , Sugar Acids/metabolism
15.
Proc Natl Acad Sci U S A ; 109(51): 20877-82, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23184990

ABSTRACT

Escherichia coli infections, a leading cause of septic shock, remain a major threat to human health because of the fatal action to endotoxin (LPS). Therapeutic attempts to neutralize endotoxin currently focus on inhibiting the interaction of the toxic component lipid A with myeloid differentiating factor 2, which forms a trimeric complex together with Toll-like receptor 4 to induce immune cell activation. The 1.73-Å resolution structure of the unique endotoxin-neutralizing protective antibody WN1 222-5 in complex with the core region shows that it recognizes LPS of all E. coli serovars in a manner similar to Toll-like receptor 4, revealing that protection can be achieved by targeting the inner core of LPS and that recognition of lipid A is not required. Such interference with Toll-like receptor complex formation opens new paths for antibody sepsis therapy independent of lipid A antagonists.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies/chemistry , Escherichia coli/metabolism , Lipopolysaccharides/chemistry , Toll-Like Receptor 4/chemistry , Animals , Antigen-Antibody Complex , Carbohydrates/chemistry , Endotoxins/metabolism , Escherichia coli Infections/metabolism , Hydrogen Bonding , Ligands , Lipids/chemistry , Mice , Mice, Inbred BALB C , Models, Chemical , Models, Molecular , Protein Binding , Shock, Septic/metabolism
16.
Innate Immun ; 18(2): 279-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21844130

ABSTRACT

The mouse monoclonal antibody (mAb) WN1 222-5 recognizes a carbohydrate epitope in the inner core region of LPS that is shared by all strains of Escherichia coli and Salmonella enterica and is able to neutralize their endotoxic activity in vitro and in vivo. Immunization of mice with mAb WN1 222-5 yielded several anti-idiotypic mAbs one of which (mAb S81-19) competitively inhibited binding of mAb WN1 222-5 to E. coli and Salmonella LPS. After immunization of rabbits with mAb S81-19, the serological responses towards LPS were characterized at intervals over two years. Whereas the serological response against the anti-idiotype developed as expected, the anti-anti-idiotypic response against LPS developed slowly and antibodies appeared after 200 d that bound to E. coli LPS of the R3 core-type and neutralized its TNF-α inducing capacity for human peripheral mononuclear cells. We describe the generation of a novel anti-idiotypic antibody that can induce LPS core-reactive antibodies upon immunization in rabbits and show that it is possible, in principle, to obtain LPS neutralizing antibodies by anti-idiotypic immunization against the mAb WN1 222-5. The mimicked epitope likely shares common determinants with the WN1 222-5 epitope, yet differences with respect to either affinity or specificity do exist, as binding to smaller oligosaccharides of the inner core was not observed.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Escherichia coli/immunology , Animals , Antibodies, Immobilized/immunology , Antigen-Antibody Reactions/drug effects , Biotinylation , Blotting, Western , Cell Fusion , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Hybridomas , Immunization , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/isolation & purification , Monocytes/drug effects , Oligosaccharides/immunology , Rabbits , Salmonella enterica/immunology , Stimulation, Chemical , Tumor Necrosis Factor-alpha/immunology
17.
Mol Cell Biol ; 32(4): 774-82, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22158965

ABSTRACT

Mannose 6-phosphate (Man6P) residues represent a recognition signal required for efficient receptor-dependent transport of soluble lysosomal proteins to lysosomes. Upon arrival, the proteins are rapidly dephosphorylated. We used mice deficient for the lysosomal acid phosphatase Acp2 or Acp5 or lacking both phosphatases (Acp2/Acp5(-/-)) to examine their role in dephosphorylation of Man6P-containing proteins. Two-dimensional (2D) Man6P immunoblot analyses of tyloxapol-purified lysosomal fractions revealed an important role of Acp5 acting in concert with Acp2 for complete dephosphorylation of lysosomal proteins. The most abundant lysosomal substrates of Acp2 and Acp5 were identified by Man6P affinity chromatography and mass spectrometry. Depending on the presence of Acp2 or Acp5, the isoelectric point of the lysosomal cholesterol-binding protein Npc2 ranged between 7.0 and 5.4 and may thus regulate its interaction with negatively charged lysosomal membranes at acidic pH. Correspondingly, unesterified cholesterol was found to accumulate in lysosomes of cultured hepatocytes of Acp2/Acp5(-/-) mice. The data demonstrate that dephosphorylation of Man6P-containing lysosomal proteins requires the concerted action of Acp2 and Acp5 and is needed for hydrolysis and removal of degradation products.


Subject(s)
Acid Phosphatase/metabolism , Isoenzymes/metabolism , Mannosephosphates/metabolism , Proteins/metabolism , Acid Phosphatase/deficiency , Acid Phosphatase/genetics , Animals , Cholesterol/metabolism , Electrophoresis, Gel, Two-Dimensional , Hepatocytes/metabolism , Isoenzymes/deficiency , Isoenzymes/genetics , Mice , Mice, Knockout , Phosphorylation , Proteins/chemistry , Tandem Mass Spectrometry , Tartrate-Resistant Acid Phosphatase , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism
18.
Glycobiology ; 21(8): 1049-59, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21543444

ABSTRACT

The structure of the antigen-binding fragment from the monoclonal antibody S64-4 in complex with a pentasaccharide bisphosphate fragment from chlamydial lipopolysaccharide has been determined by x-ray diffraction to 2.6 Å resolution. Like the well-characterized antibody S25-2, S64-4 displays a pocket formed by the residues of germline sequence corresponding to the heavy and light chain V gene segments that binds the terminal Kdo residue of the antigen; however, although S64-4 shares the same heavy chain V gene segment as S25-2, it has a different light chain V gene segment. The new light chain V gene segment codes for a combining site that displays greater affinity, different specificity, and allows a novel antigen conformation that brings a greater number of antigen residues into the combining site than possible in S25-2. Further, while antibodies in the S25-2 family use complementarity determining region (CDR) H3 to discriminate among antigens, S64-4 achieves its specificity via the new light chain V gene segment and resulting change in antigen conformation. These structures reveal an intriguing parallel strategy where two different combinations of germline-coded V gene segments can act as starting points for the generation of germline antibodies against chlamydial antigens and show how anti-carbohydrate antibodies can exploit the conformational flexibility of this class of antigens to achieve high affinity and specificity independently of CDR H3.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Chlamydia/chemistry , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Animals , Antibody Affinity , Antibody Specificity , Antigen-Antibody Complex , Carbohydrate Conformation , Chlamydia/immunology , Crystallography, X-Ray , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data
19.
Biochemistry ; 50(16): 3357-68, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21405106

ABSTRACT

The crystal structures of the antigen-binding fragment of the murine monoclonal antibody (mAb) S25-39 in the presence of several antigens representing chlamydial lipopolysaccharide (LPS) epitopes based on the bacterial sugar 3-deoxy-α-D-manno-oct-2-ulosonic acid (Kdo) have been determined at resolutions from 2.4 to 1.8 Å. The antigen-binding site of this antibody differs from the well-characterized antibody S25-2 by a single mutation away from the germline of asparagine H53 to lysine, yet this one mutation results in a significant increase in avidity across a range of antigens. A comparison of the two antibody structures reveals that the mutated Lys H53 forms additional hydrogen bonds and/or charged-residue interactions with the second Kdo residue of every antigen having two or more carbohydrate residues. Significantly, the NH53K mutation results from a single nucleotide substitution in the germline sequence common among a panel of antibodies raised against glycoconjugates containing carbohydrate epitopes of chlamydial LPS. Like S25-2, S25-39 displays significant induced fit of complementarity determining region (CDR) H3 upon antigen binding, with the unliganded structure possessing a conformation distinct from those reported earlier for S25-2. The four different observed conformations for CDR H3 suggest that this CDR has evolved to exploit the recognition potential of a flexible loop while minimizing the associated entropic penalties of binding by adopting a limited number of ordered conformations in the unliganded state. These observations reveal strategies evolved to balance adaptability and specificity in the germline antibody response to carbohydrate antigens.


Subject(s)
Binding Sites, Antibody/immunology , Chlamydia/immunology , Lipopolysaccharides/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antigens, Bacterial/immunology , Complementarity Determining Regions/immunology , Epitopes/genetics , Epitopes/immunology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Heavy Chains/genetics , Mice
20.
J Biol Chem ; 285(31): 23936-44, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20489197

ABSTRACT

GlcNAc-1-phosphotransferase is a Golgi-resident 540-kDa complex of three subunits, alpha(2)beta(2)gamma(2), that catalyze the first step in the formation of the mannose 6-phosphate (M6P) recognition marker on lysosomal enzymes. Anti-M6P antibody analysis shows that human primary macrophages fail to generate M6P residues. Here we have explored the sorting and intracellular targeting of cathepsin D as a model, and the expression of the GlcNAc-1-phosphotransferase complex in macrophages. Newly synthesized cathepsin D is transported to lysosomes in an M6P-independent manner in association with membranes whereas the majority is secreted. Realtime PCR analysis revealed a 3-10-fold higher GlcNAc-1-phosphotransferase subunit mRNA levels in macrophages than in fibroblasts or HeLa cells. At the protein level, the gamma-subunit but not the beta-subunit was found to be proteolytically cleaved into three fragments which form irregular 97-kDa disulfide-linked oligomers in macrophages. Size exclusion chromatography showed that the gamma-subunit fragments lost the capability to assemble with other GlcNAc-1-phosphotransferase subunits to higher molecular complexes. These findings demonstrate that proteolytic processing of the gamma-subunit represents a novel mechanism to regulate GlcNAc-1-phosphotransferase activity and the subsequent sorting of lysosomal enzymes.


Subject(s)
Lysosomes/enzymology , Macrophages/enzymology , Mannosephosphates/chemistry , Transferases (Other Substituted Phosphate Groups)/genetics , Animals , Biological Transport , COS Cells , Cathepsin D/chemistry , Chlorocebus aethiops , Chromatography/methods , HeLa Cells , Humans , Macrophages/cytology , Macrophages/metabolism , Models, Biological , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transferases (Other Substituted Phosphate Groups)/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...