Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Proc Jpn Acad Ser B Phys Biol Sci ; 99(6): 155-172, 2023.
Article in English | MEDLINE | ID: mdl-37331814

ABSTRACT

Catalytic antibodies possess unique features capable of both recognizing and enzymatically degrading antigens. Therefore, they are more beneficial than monoclonal antibodies (mAbs). Catalytic antibodies exhibit the ability to degrade peptides, antigenic proteins, DNA, and physiologically active molecules. However, they have a significant drawback in terms of their production. The production of a desired catalytic antibody has extensive costs, in terms of time and effort. We herein describe an evolutionary method to produce a desired catalytic antibody via conversion of a general antibody by the deletion of Pro95, which resides in complementarity-determining region-3. As over thousands of mAbs have been produced since 1975, using the novel technology discussed herein, the catalytic feature cleaving the antigen can be conferred to the mAb. In this review article, we discussed in detail not only the role of Pro95 but also the unique features of the converted catalytic antibodies. This technique will accelerate research on therapeutic application of catalytic antibodies.


Subject(s)
Antibodies, Catalytic , Antibodies, Catalytic/chemistry , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism
2.
Int J Mol Sci ; 23(22)2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36430828

ABSTRACT

A catalytic antibody has multiple functions compared with a monoclonal antibody because it possesses unique features to digest antigens enzymatically. Therefore, many catalytic antibodies, including their subunits, have been produced since 1989. The catalytic activities often depend on the preparation methods and conditions. In order to elicit the high catalytic activity of the antibodies, the most preferable methods and conditions, which can be generally applicable, must be explored. Based on this view, systematic experiments using two catalytic antibody light chains, #7TR and H34, were performed by varying the purification methods, pH, and chemical reagents. The experimental results obtained by peptidase activity tests and kinetic analysis, revealed that the light chain's high catalytic activity was observed when it was prepared under a basic condition. These data imply that a small structural modulation of the catalytic antibody occurs during the purification process to increase the catalytic activity while the antigen recognition ability is kept constant. The presence of NaCl enhanced the catalytic activity. When the catalytic light chain was prepared with these preferable conditions, #7TR and H34 hugely enhanced the degradation ability of Amyloid-beta and PD-1 peptide, respectively.


Subject(s)
Antibodies, Catalytic , Antibodies, Catalytic/chemistry , Kinetics , Antigens , Immunoglobulin Light Chains , Antibodies, Monoclonal
3.
Langmuir ; 37(50): 14760-14766, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34889092

ABSTRACT

Transformable double hydrophilic block copolymer assemblies are valid as a biocompatible smart macromolecular system. The molecular mechanisms in the spontaneous assembly of double zwitterionic diblock copolymers composed of a poly(carboxybetaine methacrylate) (PCB2) and a poly(sulfobetaine methacrylate) (PSB4) chains (PCB2-b-PSB4) were investigated by the modulation of the aggregates in response to nondetergent zwitterions. The PCB2-b-PSB4 diblock copolymers with a high degree of polymerization PSB4 block produced aggregates in salt-free water through "zwitterion-specific" interactions. The PCB2-b-PSB4 aggregates were dissociated by the addition of nondetergent sulfobetaine (SB4) and carboxybetaine (CB2) molecules, while the aggregates showed different aggregation modulation processes for SB4 and CB2. Zwitterions with different charged groups from SB4 and CB2, glycine and taurine, hardly disrupted the PCB2-b-PSB4 aggregates. The PCB2-b-PSB4 aggregate modulation efficiency of SBs associated with the intercharge hydrocarbon spacer length (CSL) rather than the symmetry with the SB in the PSB chain. These zwitterion-specific modulation behaviors were rationalized based on the nature of zwitterions including partial charge density, dipole moment, and hydrophobic interactions depending on the charged groups and CSL.


Subject(s)
Methacrylates , Polymers , Hydrophobic and Hydrophilic Interactions , Polymerization
4.
Photochem Photobiol Sci ; 19(1): 29-33, 2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31932832

ABSTRACT

We demonstrate amyloid fibril (AF) decomposition induced by NIR-active upconversion nanoparticles complexed with photosensitisers. The process is triggered by upconversion, which initiates a photochemical reaction cascade that culminates in the generation of the highly reactive singlet-oxygen product 1O2 close to the amyloid superstructures, resulting in AF decomposition.


Subject(s)
Amyloid/antagonists & inhibitors , Nanoparticles/chemistry , Photosensitizing Agents/pharmacology , Amyloid/metabolism , Humans , Infrared Rays
5.
FASEB J ; 31(4): 1668-1677, 2017 04.
Article in English | MEDLINE | ID: mdl-28096233

ABSTRACT

Issues regarding the structural diversity (heterogeneity) of an antibody molecule have been the subject of discussion along with the development of antibody drugs. Research on heterogeneity has been extensive in recent years, but no clear solution has been reached. Heterogeneity is also observed in catalytic antibody κ light chains (CLs). In this study, we investigated how the constant region domain of CLs concerns structural diversity because it is a simple and good example for elucidating heterogeneity. By means of cation-exchange chromatography, SDS-PAGE, and 2-dimensional electrophoresis for the CL, multimolecular forms consisting of different electrical charges and molecular sizes coexisted in the solution, resulting in the similar heterogeneity of the full length of CLs. The addition of copper ion could cause the multimolecular forms to change to monomolecular forms. Copper ion contributed greatly to the enrichment of the dimer form of CL and the homogenization of the differently charged CLs. Two molecules of the CL protein bound one copper ion. The binding affinity of the ion was 48.0 µM-1 Several divalent metal ions were examined, but only zinc showed a similar effect.-Hifumi, E., Taguchi, H., Kato, R., Uda, T. Role of the constant region domain in the structural diversity of human antibody light chains.


Subject(s)
Immunoglobulin Constant Regions/chemistry , Immunoglobulin Light Chains/chemistry , Copper/pharmacology , Genetic Heterogeneity , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Constant Regions/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Protein Binding , Protein Domains , Protein Multimerization , Static Electricity , Zinc/pharmacology
6.
FASEB J ; 30(2): 895-908, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26527062

ABSTRACT

Along with the development of antibody drugs and catalytic antibodies, the structural diversity (heterogeneity) of antibodies has been given attention. For >20 yr, detailed studies on the subject have not been conducted, because the phenomenon presents many difficult and complex problems. Structural diversity provides some (or many) isoforms of an antibody distinguished by different charges, different molecular sizes, and modifications of amino acid residues. For practical use, the antibody and the subunits must have a defined structure. In recent work, we have found that the copper (Cu) ion plays a substantial role in solving the diversity problem. In the current study, we used several catalytic antibody light chains to examine the effect of the Cu ion. In all cases, the different electrical charges of the molecule converged to a single charge, giving 1 peak in cation-exchange chromatography, as well as a single spot in 2-dimensional gel electrophoresis. The Cu-binding site was investigated by using mutagenesis, ultraviolet-visible spectroscopy, atomic force microscope analysis, and molecular modeling, which suggested that histidine and cysteine residues close to the C-terminus are involved with the binding site. The constant region domain of the antibody light chain played an important role in the heterogeneity of the light chain. Our findings may be a significant tool for preparing a single defined, not multiple, isoform structure.


Subject(s)
Antibodies, Catalytic/chemistry , Copper/chemistry , Immunoglobulin Light Chains/chemistry , Antibodies, Catalytic/isolation & purification , Binding Sites, Antibody , Humans , Immunoglobulin Light Chains/isolation & purification
7.
FASEB J ; 29(6): 2347-58, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25713031

ABSTRACT

Catalytic antibodies have exhibited interesting functions against some infectious viruses such as HIV, rabies virus, and influenza virus in vitro as well as in vivo. In some cases, a catalytic antibody light chain takes on several structures from the standpoint of molecular size (monomer, dimer, etc.) and/or isoelectronic point. In this study, we prepared a monomeric 23D4 light chain by mutating the C-terminal Cys to Ala of the wild-type. The mutated 23D4 molecule took a simple monomeric form, which could hydrolyze synthetic 4-methyl-coumaryl-7-amide substrates and a plasmid DNA. Because the monomeric 23D4 light chain suppressed the infection of influenza virus A/Hiroshima/37/2001 in an in vitro assay, the corresponding experiments were conducted in vivo, after the virus strain (which was taken from a human patient) was successfully adapted into BALB/cN Sea mice. In the experiments, a mixture of the monomeric 23D4 and the virus was nasally administered 1) with preincubation and 2) without preincubation. As a result, the monomeric 23D4 clearly exhibited the ability to suppress the influenza virus infection in both cases, indicating a potential drug for preventing infection of the influenza A virus.


Subject(s)
Antibodies, Catalytic/immunology , Antiviral Agents/immunology , Immunoglobulin Light Chains/immunology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Catalytic/genetics , Antibodies, Catalytic/metabolism , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Blotting, Western , Coumarins/immunology , Coumarins/metabolism , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Mice, Inbred BALB C , Microscopy, Atomic Force , Mutation , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Substrate Specificity
8.
Anal Bioanal Chem ; 408(22): 5963-71, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27173395

ABSTRACT

Recently, point of care testing (POCT) used for diagnosis of influenza infection has a problem showing false negative diagnosis because of the low sensitivity. We would like to report detection of influenza virus A (H1N1) by an immunosensor based on electrochemiluminescence (ECL) that uses an immunoliposome encapsulating tris(2,2'-bipyridyl)ruthenium(II) complex. By using the sensor, we could detect the virus that competed with hemagglutinin (HA) peptide immobilized on self-assembled monolayers (SAMs) in immunoreaction of the antibody bound on the surface of liposome. The HA peptide was 19 mer (TGLRNGITNKVNSVIEKAA). We demonstrated great improvement of sensitivity and accuracy by introducing binary SAMs instead of mono SAMs. The binary SAMs was prepared from 3,3'-dithiodipropionic acid and 1-hexanethiol. Use of the binary SAMs enabled to increase the SAMs coverage on Au electrode; the fact was confirmed by observation of the cathodic desorption currents. By using such an electrode, first the detection method of BSA was optimized to lower ECL background signal. Then we applied the method to the detection of influenza virus. We could successfully detect the virus with higher sensitivity compared with that by POCT and ELISA. The detection range was from a concentration of 2.7 × 10(2) to 2.7 × 10(3) PFU/mL.


Subject(s)
Biosensing Techniques/methods , Influenza A Virus, H1N1 Subtype/isolation & purification , Liposomes/chemistry , Luminescent Measurements/methods , Organometallic Compounds/chemistry , Animals , Antibodies, Immobilized/chemistry , Cattle , Hemagglutinins/chemistry , Humans , Immobilized Proteins/chemistry , Immunoassay/methods , Influenza, Human/diagnosis , Influenza, Human/virology , Peptides/chemistry , Sensitivity and Specificity , Serum Albumin, Bovine/analysis
9.
J Biol Chem ; 288(27): 19558-68, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23677996

ABSTRACT

Human antibody light chains belonging to subgroup II of germ line genes were amplified by a seminested PCR technique using B-lymphocytes taken from a human adult infected with influenza virus. Each gene of the human light chains was transferred into the Escherichia coli system. The recovered light chain was highly purified using a two-step purification system. Light chain 22F6 showed interesting catalytic features. The light chain cleaved a peptide bond of synthetic peptidyl-4-methyl-coumaryl-7-amide (MCA) substrates, such as QAR-MCA and EAR-MCA, indicating amidase activity. It also hydrolyzed a phosphodiester bond of both DNA and RNA. From the analysis of amino acid sequences and molecular modeling, the 22F6 light chain possesses two kinds of active sites as amidase and nuclease in close distances. The 22F6 catalytic light chain could suppress the infection of influenza virus type A (H1N1) of Madin-Darby canine kidney cells in an in vitro assay. In addition, the catalytic light chain clearly inhibited the infection of the influenza virus of BALB/c mice via nasal administration in an in vivo assay. In the experiment, the titer in the serum of the mice coinfected with the 22F6 light chain and H1N1 virus became considerably lowered compared with that of 22F6-non-coinfected mice. Note that the catalytic light chain was prepared from human peripheral lymphocyte and plays an important role in preventing infection by influenza virus. Considering the fact that the human light chain did not show any acute toxicity for mice, our procedure developed in this study must be unique and noteworthy for developing new drugs.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , B-Lymphocytes/immunology , Immunoglobulin Light Chains , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/drug therapy , Adult , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Base Sequence , Dogs , Female , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin Light Chains/pharmacology , Madin Darby Canine Kidney Cells , Male , Mice , Molecular Sequence Data , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology
10.
Sci Rep ; 14(1): 12184, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806597

ABSTRACT

Catalytic antibodies possess a dual function that enables both antigen recognition and degradation. However, their time-consuming preparation is a significant drawback. This study developed a new method for quickly converting mice monoclonal antibodies into catalytic antibodies using site-directed mutagenesis. Three mice type monoclonal antibodies targeting hemagglutinin molecule of influenza A virus could be transformed into the catalytic antibodies by deleting Pro95 in CDR-3 of the light chain. No catalytic activity was observed for monoclonal antibodies and light chains. In contrast, the Pro95-deleted light chains exhibited a catalytic activity to cleave the antigenic peptide including the portion of conserved region of hemagglutinin molecule. The affinity of the Pro95-deleted light chains to the antigen increased approximately 100-fold compared to the wild-type light chains. In the mutants, three residues (Asp1, Ser92, and His93) come closer to the appropriate position to create the catalytic site and contributing to the enhancement of both catalytic function and immunoreactivity. Notably, the Pro95-deleted catalytic light chains could suppress influenza virus infection in vitro assay, whereas the parent antibody and the light chain did not. This strategy offers a rapid and efficient way to create catalytic antibodies from existing antibodies, accelerating the development for various applications in diagnostic and therapeutic applications.


Subject(s)
Antibodies, Catalytic , Antibodies, Monoclonal , Animals , Mice , Antibodies, Monoclonal/immunology , Antibodies, Catalytic/metabolism , Antibodies, Catalytic/immunology , Antibodies, Catalytic/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Mutagenesis, Site-Directed , Influenza A virus/immunology , Catalytic Domain , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin Light Chains/metabolism , Antibodies, Viral/immunology , Mice, Inbred BALB C
11.
FASEB J ; 26(4): 1607-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22205784

ABSTRACT

The ultimate goal of catalytic antibody research is to develop new patient therapies that use the advantages offered by human catalytic antibodies. The establishment of a high-throughput method for obtaining valuable candidate catalytic antibodies must be accelerated to achieve this objective. In this study, based on our concept that we can find antibody light chains with a high probability of success if they include a serine protease-like catalytic triad composed of Ser, His, and Asp on a variable region of the antibody structure, we amplified and cloned DNAs encoding human antibody light chains from germline genes of subgroup II by seminested PCR using two primer sets designed for this purpose. Seven DNA fragments encoding light chains in 17 clones were derived from germline gene A18b, 6 DNA fragments from A3/A19, 2 DNA fragments from A17, and a clone DNA fragment from A5 and O11/O1. All light chains expressed in Escherichia coli and highly purified under nondenaturing conditions exhibited amidolytic activity against synthetic peptides. Some of the light chains exhibited unique features that suppressed the infectious activity of the rabies virus. Furthermore, the survival rate of mice in which a lethal level of the rabies virus was coinoculated directly into the brain with light chain 18 was significantly improved. In the case of humans, these results demonstrate that high-throughput selection of light chains possessing catalytic functions and specificity for a target molecule can be attained from a light-chain DNA library amplified from germline genes belonging to subgroup II.


Subject(s)
Antibodies, Catalytic/immunology , Immunoglobulin Light Chains/genetics , Polymerase Chain Reaction/methods , Algorithms , Amino Acid Sequence , Animals , Animals, Suckling , Antibodies, Catalytic/chemistry , Antibodies, Catalytic/genetics , DNA/genetics , DNA/metabolism , Germ Cells , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Rabies virus/immunology , Sequence Alignment , Survival Rate
12.
Nat Commun ; 14(1): 7807, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38065949

ABSTRACT

Overexpression of antibody light chains in small plasma cell clones can lead to misfolding and aggregation. On the other hand, the formation of amyloid fibrils from antibody light chains is related to amyloidosis. Although aggregation of antibody light chain is an important issue, atomic-level structural examinations of antibody light chain aggregates are sparse. In this study, we present an antibody light chain that maintains an equilibrium between its monomeric and tetrameric states. According to data from X-ray crystallography, thermodynamic and kinetic measurements, as well as theoretical studies, this antibody light chain engages in 3D domain swapping within its variable region. Here, a pair of domain-swapped dimers creates a tetramer through hydrophobic interactions, facilitating the revelation of the domain-swapped structure. The negative cotton effect linked to the ß-sheet structure, observed around 215 nm in the circular dichroism (CD) spectrum of the tetrameric variable region, is more pronounced than that of the monomer. This suggests that the monomer contains less ß-sheet structures and exhibits greater flexibility than the tetramer in solution. These findings not only clarify the domain-swapped structure of the antibody light chain but also contribute to controlling antibody quality and advancing the development of future molecular recognition agents and drugs.


Subject(s)
Amyloidosis , Immunoglobulin Light Chains , Humans , Amyloid/chemistry , Crystallography, X-Ray , Thermodynamics
13.
Sci Rep ; 12(1): 19185, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36357546

ABSTRACT

The cleavage reactions of catalytic antibodies are mediated by a serine protease mechanism involving a catalytic triad composed of His, Ser, and Asp residues, which reside in the variable region. Recently, we discovered a catalytic antibody, H34 wild type (H34wt), that is capable of enzymatically cleaving an immune-check point PD-1 peptide and recombinant PD-1; however, H34wt does not contain His residues in the variable region. To clarify the reason behind the catalytic features of H34wt and the amino acid residues involved in the catalytic reaction, we performed site-directed mutagenesis focusing on the amino acid residues involved in the cleavage reaction, followed by catalytic activity tests, immunological reactivity evaluation, and molecular modeling. The results revealed that the cleavage reaction by H34wt proceeds through the action of a new catalytic site composed of Arg, Thr, and Gln. This new scheme differs from that of the serine protease mechanism of catalytic antibodies.


Subject(s)
Antibodies, Catalytic , Catalytic Domain , Antibodies, Catalytic/chemistry , Antibodies, Catalytic/metabolism , Amino Acid Sequence , Programmed Cell Death 1 Receptor , Immunoglobulin Light Chains/metabolism , Serine Endopeptidases/metabolism , Amino Acids
14.
J Am Chem Soc ; 133(38): 15015-24, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21861493

ABSTRACT

It has long been an important issue to produce a catalytic antibody that possesses the ability to lose the infectivity of a bacteria or virus. The monoclonal antibody JN1-2 was generated using a synthetic peptide (TGLRNGITNKVNSVIEKAA) conjugated with human IgG. The peptide sequence includes the conserved region of the hemagglutinin molecule (HA(1) and HA(2) domains), which locates on the envelope of the influenza virus and plays an important role in influenza A virus infection. The monoclonal antibody specifically reacted with the HA2 domain, not only of H2 but also of an H1 strain of the H1N1 subtype (H1 strain). The heavy chain (JN1-2-H) isolated from the parent antibody showed catalytic activity cleaving the above antigenic peptide with very high turnover (kcat = 26 min(-1)), and it could slowly degrade the recombinant HA(2) domain by the catalytic function. Interestingly, the heavy chain exhibited the ability to reduce the infectivity of type A H1N1 but not type B, indicating specificity to type A. This characteristic monoclonal catalytic antibody heavy chain could suppress the infection of the influenza virus in vitro assays.


Subject(s)
Antibodies, Monoclonal/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Oligopeptides/chemistry , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Biochemistry , Catalysis , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Models, Molecular , Oligopeptides/chemical synthesis , Oligopeptides/immunology
15.
RSC Chem Biol ; 2(1): 220-229, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-34458785

ABSTRACT

Programmed cell death 1 (PD-1) is an immune checkpoint molecule regulating T-cell function. Preventing PD-1 binding to its ligand PD-L1 has emerged as an important tool in immunotherapy. Here, we describe a unique human catalytic antibody light chain, H34, which mediates enzymatic degradation of human PD-1 peptides and recombinant human PD-1 protein and thus functions to prevent the binding of PD-1 with PD-L1. H34 degraded one half of the PD-1 molecules within about 6 h under the experimental conditions. Investigating the acquisition of the catalytic function by H34, which belongs to subgroup I and lacks a Pro95 residue in CDR-3, revealed the importance of this sequence, as a Pro95-reconstituted mutant (H34-Pro95(+)) exhibited very little catalytic activity to cleave PD-1. Interestingly, EDTA inhibited the catalytic activity of H34, which could work as a metallo-protease. Zn2+ or Co2+ ions may work as a cofactor. It is meaningfull that H34 was obtained from the human antibody gene taken from a healthy volunteer, suggesting that we potentially have such unique molecules in our body.

16.
Sci Adv ; 6(13): eaay6441, 2020 03.
Article in English | MEDLINE | ID: mdl-32232151

ABSTRACT

Over thousands of monoclonal antibodies (mAbs) have been produced so far, and it would be valuable if these mAbs could be directly converted into catalytic antibodies. We have designed a system to realize the above concept by deleting Pro95, a highly conserved residue in CDR-3 of the antibody light chain. The deletion of Pro95 is a key contributor to catalytic function of the light chain. The S35 and S38 light chains have identical amino acid sequences except for Pro95. The former, with Pro95 did not show any catalytic activity, whereas the latter, without Pro95, exhibited peptidase activity. To verify the generality of this finding, we tested another light chain, T99wt, which had Pro95 and showed little catalytic activity. In contrast, a Pro95-deleted mutant enzymatically degraded the peptide substrate and amyloid-beta molecule. These two cases demonstrate the potential for a new method of creating catalytic antibodies from the corresponding mAbs.


Subject(s)
Algorithms , Antibodies, Catalytic/chemistry , Antibodies, Monoclonal/chemistry , Models, Molecular , Amino Acid Sequence , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Antibodies, Catalytic/genetics , Antibodies, Catalytic/metabolism , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Catalysis , Chromatography, High Pressure Liquid , Hydrolysis , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Mutation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation , Protein Multimerization , Proteolysis , Recombinant Proteins
17.
FASEB Bioadv ; 1(2): 93-104, 2019 Feb.
Article in English | MEDLINE | ID: mdl-32123823

ABSTRACT

Since the discovery of a natural catalytic antibody in 1989, many catalytic antibodies targeting peptides, nucleotides, virus and bacterial proteins, and many molecules have been prepared. Although catalytic antibodies should have features superior to non-catalytic monoclonal antibodies, the reports on catalytic antibodies are far fewer than those on normal (non-catalytic) antibodies. Nowadays, we can obtain any monoclonal antibody we want, which is not the case for catalytic antibodies. To overcome this drawback of catalytic antibodies, much basic research must be done. As one way to attain this purpose, we have been making a protein bank of human antibody light chains, in which the light chains were expressed, purified, and stored for use in screening against many kinds of antigen, to then get clues to introducing a catalytic function in normal antibodies. As the number of stored light chains in the above protein bank has reached the hundreds, in this study, we screened them against amyloid-beta (Aß), which is well-known as one of the molecules causing Alzheimer's disease. We found two interesting light chains, #7TR and #7GY. The former could degrade both a fluorescence resonance energy transfer-Aß substrate and Aß1-40 full peptide. In contrast, #7GY, whose sequence is identical to that of #7TR except for the amino acids at the 29th and 30th positions, did not degrade the FRET-Aß substrate at all. By using a synthetic substrate, Arg-pNA, the difference between the chemical features of the two light chains was investigated in detail. In addition, we found that the presence of Zn(II) ion hugely influenced the catalytic activity of the #7TR light chain but not #7GY. Through these facts and the discussion, we propose one of the clues to how to put a catalytic function in a normal (non-catalytic) antibody.

18.
Anal Chem ; 80(11): 4020-5, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18447321

ABSTRACT

An immunoliposome (80 nm in diameter) encapsulating a Ru complex with two aminobutyl moieties was prepared to detect the presence of hemagglutinin molecules, which play an important role in influenza virus infection. The highly sensitive detection was accomplished by electrochemiluminescence (ECL) from the Ru complex adsorbed onto Au electrodes after competitive immunoreactions. This method clarified that the adsorption of the Ru complex onto the electrode was an important factor in obtaining high sensitivity. Optimization of the analytical conditions enabled determination of the hemagglutinin molecules of the influenza virus in the concentration range of 3 x 10(-14) (6 x 10(-19) mol/50 microL sample) to 2 x 10(-12) g/mL. The sensitivity was far superior to that obtained by conventional ELISA as well as to that obtained by biosensors and reported thus far.


Subject(s)
Biosensing Techniques/instrumentation , Hemagglutinins/analysis , Immunoassay/methods , Liposomes/immunology , Luminescent Measurements/methods , Organometallic Compounds/immunology , Orthomyxoviridae/chemistry , Ruthenium Compounds/immunology , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Calibration , Electrochemistry , Electrodes , Gold/chemistry , Hemagglutinins/immunology , Molecular Sequence Data , Organometallic Compounds/chemistry , Orthomyxoviridae/immunology , Orthomyxoviridae/isolation & purification , Peptides/chemistry , Ruthenium Compounds/chemistry , Sensitivity and Specificity
19.
Biotechnol Bioeng ; 100(4): 634-43, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18306426

ABSTRACT

Helicobacter pylori (H. pylori) is a bacteria that is well known as the principal cause of chronic gastritis and peptic ulcer disease in humans. Because no effective vaccine has yet been established, we designed a new biomolecule as a vaccination antigen capable of preventing the infection of H. pylori. The designed biomolecule involves a 138 stretch (aa 201-aa 338 of beta-subunit of H. pylori urease), which is the functionally important region for urease activity. The region was expressed as a recombinant protein, called UREB138. The therapeutic vaccination was performed using UREB138 in mice persistently infected with H. pylori. The subcutaneous administration of UREB138 remarkably reduced the number of bacteria in the mice stomach compared with the control. Immunization with UREB138 enhanced the urease-specific IgA and IgG1 in the serum. Immunohistochemical staining for IgA in gastric mucosa showed that the number of mice positively stained with IgA was significantly higher in UREB138-immunized mice than in control mice. Furthermore, the expression of interferon-gamma mRNA in the gastric tissues with eradicated bacteria was higher than in the non-eradicated group. The combination of Th1- and Th2-mediated immunity plays a role in reducing the colonization of bacterial numbers of H. pylori.


Subject(s)
Antigens/therapeutic use , Helicobacter Infections/therapy , Helicobacter pylori , Immunization , Urease/antagonists & inhibitors , Urease/immunology , Animals , Antigens/administration & dosage , Antigens/genetics , Disease Models, Animal , Enzyme Inhibitors/immunology , Female , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gene Expression/immunology , Helicobacter Infections/immunology , Helicobacter pylori/enzymology , Helicobacter pylori/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Injections, Subcutaneous , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Inbred Strains , Protein Subunits/genetics , Protein Subunits/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
20.
J Med Microbiol ; 56(Pt 6): 847-853, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17510273

ABSTRACT

Helicobacter pylori has to counteract acidity during colonization in the stomach. The most important region for the enzymic activity of H. pylori urease, consisting of 138 aa (ureB138), was determined by a comparison of the homology of amino acid sequences, and a structural analysis, between urease of H. pylori and various other species. This region was expressed in Escherichia coli as a fusion protein with glutathione S-transferase (GST), which was cleaved by PreScission protease between the GST moiety and ureB138. The ureB138 protein was then purified by gel filtration. The polyclonal antibody (pAb) induced by immunization with the purified ureB138 could suppress urease activity by about 50 %, while the pAb against the H. pylori urease did not show any inhibitory effect at all. Immunohistochemical analysis indicated that the ureB138-specific pAb specifically recognized the H. pylori infecting human gastric tissues. The effects of vaccination of recombinant ureB138 against infection by this organism were also examined. Specific IgG and IgA antibodies against H. pylori urease were induced in the serum of mice immunized with ureB138. A reduction in the number of colonizing H. pylori was observed in mice treated with ureB138 compared to ones treated with BSA and infection control mice. In the protected mice, severe gastritis characterized by marked infiltration of mononuclear cells was noted compared with the gastritis observed in unprotected mice. Immunohistochemical staining for IgA in gastric mucosa showed that the number of mice positively stained with IgA was significantly higher in ureB138-vaccinated mice than in non-vaccinated mice. This indicates that local IgA antibody and severe post-immunization gastritis correlate well with the protection of mice against H. pylori infection.


Subject(s)
Bacterial Vaccines/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Urease/immunology , Animals , Antibodies, Bacterial/immunology , Bacterial Vaccines/genetics , Colony Count, Microbial , Disease Models, Animal , Escherichia coli/genetics , Female , Gene Expression , Helicobacter Infections/immunology , Histocytochemistry , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy , Protein Subunits/genetics , Protein Subunits/immunology , Stomach/immunology , Stomach/microbiology , Urease/antagonists & inhibitors , Urease/genetics , Urease/isolation & purification , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL