Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 479(2): 245-252, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27638305

ABSTRACT

A direct acting anti-Ebola agent is needed. VP40, a conserved protein across Ebolavirus (EBOV) species has several pivotal roles in the virus life cycle. Inhibition of VP40 functions would lessen the virion integrity and interfere with the viral assembly, budding, and spread. In this study, cell penetrable human scFvs (HuscFvs) that bound to EBOV VP40 were produced by phage display technology. Gene sequences coding for VP40-bound-HuscFvs were subcloned from phagemids into protein expression plasmids downstream to a gene of cell penetrating peptide, i.e., nonaarginine (R9). By electron microscopy, transbodies from three clones effectively inhibited egress of the Ebola virus-like particles from human hepatic cells transduced with pseudo-typed-Lentivirus particles carrying EBOV VP40 and GP genes. Computerized simulation indicated that the effective HuscFvs bound to multiple basic residues in the cationic patch of VP40 C-terminal domain which are important in membrane-binding for viral matrix assembly and virus budding. The transbodies bound also to VP40 N-terminal domain and L domain peptide encompassed the PTAPPEY (WW binding) motif, suggesting that they might confer VP40 function inhibition through additional mechanism(s). The generated transbodies are worthwhile tested with authentic EBOV before developing to direct acting anti-Ebola agent for preclinical and clinical trials.


Subject(s)
Ebolavirus/drug effects , Single-Chain Antibodies/pharmacology , Viral Matrix Proteins/immunology , Virus Release/drug effects , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/ultrastructure , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Ebolavirus/physiology , Ebolavirus/ultrastructure , Host-Pathogen Interactions , Humans , Liver Neoplasms/pathology , Liver Neoplasms/ultrastructure , Liver Neoplasms/virology , Microscopy, Electron, Scanning , Models, Molecular , Peptide Library , Protein Binding , Protein Domains , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Virion/drug effects , Virion/physiology , Virion/ultrastructure , Virus Release/physiology
2.
Protein Eng Des Sel ; 27(5): 135-44, 2014 May.
Article in English | MEDLINE | ID: mdl-24760832

ABSTRACT

Two LipL32-specific mouse monoclonal antibodies (mAbLPF1 and mAbLPF2) which neutralized Leptospira-mediated hemolysis in vitro and rescued hamsters from lethal Leptospira infection were produced. In this communication, locations and characteristics of the protective epitopes of the mAbs were studied by using a truncated LipL32 recombinant protein based-immunoassay and phage consensus mimotope identification and multiple alignments. The mAbLPF1 epitope consisted of P243, L244, I245, H246, L252 and Q253 on the LipL32 protein; it is mapped on the surface-exposed region of non-continuous ß13-turn and C-terminal amphipathic α6 helix with hydrophobic patch, contributing to phospholipid/host cell adhesion and membrane insertion on one side, and hydrophilic, acidic and basic amino acid residues on another side. The epitope peptide of the mAbLPF2 is linear 122PEEKSMPHW130 and located on surface-exposed α1 and α2 between ß7 and ß8 that bound to several host constituents. Both epitopes are highly conserved among the pathogenic and intermediately pathogenic Leptospira spp. and are absent from the LipL32 superfamily proteins of other microorganisms. This study not only enlightens the molecular mechanisms of the therapeutic mAbLPF1 and mAbLPF2, but also elaborates the potential of the two LipL32 regions as diagnostic and vaccine targets for leptospirosis.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Epitopes/immunology , Epitopes/therapeutic use , Lipoproteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibody Specificity , Bacterial Outer Membrane Proteins/chemistry , Cricetinae , Epitope Mapping , Epitopes/chemistry , Leptospira/immunology , Leptospira/physiology , Leptospirosis/drug therapy , Lipoproteins/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary
3.
Antiviral Res ; 100(1): 226-37, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23928258

ABSTRACT

Currently, there is a need of new anti-influenza agents that target influenza virus proteins other than ion channel M2 and neuraminidase. Non-structural protein-1 (NS1) is a highly conserved multifunctional protein which is indispensable for the virus replication cycle. In this study, fully human single chain antibody fragments (HuScFv) that bound specifically to recombinant and native NS1 were produced from three huscfv-phagemid transformed Escherichia coli clones (nos. 3, 10 and 11) selected from a human ScFv phage display library. Western blot analysis, mimotope searching/epitope identification, homology modeling/molecular docking and phage mimotope ELISA inhibition indicated that HuScFv of clone no. 3 reacted with NS1 R domain important for host innate immunity suppression; HuScFv of clone nos. 10 and 11 bound to E domain sites necessary for NS1 binding to the host eIF4GI and CPSF30, respectively. The HuScFv of all clones could enter the influenza virus infected cells and interfered with the NS1 activities leading to replication inhibition of viruses belonging to various heterologous A subtypes and type B by 2-64-fold as semi-quantified by hemagglutination assay. Influenza virus infected cells treated with representative HuScFv (clone 10) had up-expression of IRF3 and IFN-ß genes by 14.75 and 4.95-fold, respectively, in comparison with the controls, indicating that the antibodies could restore the host innate immune response. The fully human single chain antibodies have high potential for developing further as a safe (adjunctive) therapeutic agent for mitigating, if not abrogating, severe symptoms of influenza.


Subject(s)
Antibodies, Viral/pharmacology , Down-Regulation/drug effects , Influenza A virus/drug effects , Influenza, Human/virology , Single-Chain Antibodies/pharmacology , Viral Nonstructural Proteins/immunology , Virus Replication/drug effects , Animals , Chick Embryo , Humans , Influenza A virus/classification , Influenza A virus/immunology , Influenza A virus/physiology , Influenza, Human/drug therapy , Viral Nonstructural Proteins/genetics
4.
Virol J ; 10: 148, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23672256

ABSTRACT

BACKGROUND: Novel effective anti-influenza agent that tolerates influenza virus antigenic variation is needed. Highly conserved influenza virus M2 protein has multiple pivotal functions including ion channel activity for vRNP uncoating, anti-autophagy and virus assembly, morphogenesis and release. Thus, M2 is an attractive target of anti-influenza agents including small molecular drugs and specific antibodies. METHODS: Fully human monoclonal single chain antibodies (HuScFv) specific to recombinant and native M2 proteins of A/H5N1 virus were produced from huscfv-phagemid transformed E. coli clones selected from a HuScFv phage display library using recombinant M2 of clade 1 A/H5N1 as panning antigen. The HuScFv were tested for their ability to inhibit replication of A/H5N1 of both homologous and heterologous clades. M2 domains bound by HuScFv of individual E. coli clones were identified by phage mimotope searching and computerized molecular docking. RESULTS: HuScFv derived from four huscfv-phagemid transformed E. coli clones (no. 2, 19, 23 and 27) showed different amino acid sequences particularly at the CDRs. Cells infected with A/H5N1 influenza viruses (both adamantane sensitive and resistant) that had been exposed to the HuScFv had reduced virus release and intracellular virus. Phage peptide mimotope search and multiple alignments revealed that conformational epitopes of HuScFv2 located at the residues important for ion channel activity, anti-autophagy and M1 binding; epitopic residues of HuScFv19 located at the M2 amphipathic helix and cytoplasmic tail important for anti-autophagy, virus assembly, morphogenesis and release; epitope of HuScFv23 involved residues important for the M2 activities similar to HuScFv2 and also amphipathic helix residues for viral budding and release while HuScFv27 epitope spanned ectodomain, ion channel and anti-autophagy residues. Results of computerized homology modelling and molecular docking conformed to the epitope identification by phages. CONCLUSIONS: HuScFv that bound to highly conserved epitopes across influenza A subtypes and human pathogenic H5N1clades located on different functional domains of M2 were produced. The HuScFv reduced viral release and intracellular virus of infected cells. While the molecular mechanisms of the HuScFv await experimental validation, the small human antibody fragments have high potential for developing further as a safe, novel and mutation tolerable anti-influenza agent especially against drug resistant variants.


Subject(s)
Antibodies, Viral/immunology , Antiviral Agents/isolation & purification , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/physiology , Single-Chain Antibodies/immunology , Viral Matrix Proteins/immunology , Virus Replication , Animals , Antibodies, Viral/isolation & purification , Antibodies, Viral/metabolism , Antiviral Agents/metabolism , Chick Embryo , Escherichia coli/genetics , Gene Expression , Genetic Vectors , Humans , Peptide Library , Protein Binding , Single-Chain Antibodies/isolation & purification , Single-Chain Antibodies/metabolism , Viral Matrix Proteins/metabolism
5.
PLoS One ; 7(11): e49254, 2012.
Article in English | MEDLINE | ID: mdl-23145135

ABSTRACT

NS5B is pivotal RNA dependent RNA polymerase (RdRp) of HCV and NS5B function interfering halts the virus infective cycle. This work aimed to produce cell penetrable humanized single domain antibodies (SdAb; VH/V(H)H) that interfere with the RdRp activity. Recombinant NS5BΔ55 of genotype 3a HCV with de novo RNA synthetic activity was produced and used in phage biopanning for selecting phage clones that displayed NS5BΔ55 bound VH/V(H)H from a humanized-camel VH/V(H)H display library. VH/V(H)H from E. coli transfected with four selected phage clones inhibited RdRp activity when tested by ELISA inhibition using 3'di-cytidylate 25 nucleotide directed in vitro RNA synthesis. Deduced amino acid sequences of two clones showed V(H)H hallmark and were designated V(H)H6 and V(H)H24; other clones were conventional VH, designated VH9 and VH13. All VH/V(H)H were linked molecularly to a cell penetrating peptide, penetratin. The cell penetrable VH9, VH13, V(H)H6 and V(H)H24 added to culture of Huh7 cells transfected with JHF-1 RNA of genotype 2a HCV reduced the amounts of RNA intracellularly and in culture medium implying that they inhibited the virus replication. VH/V(H)H mimotopes matched with residues scattered on the polymerase fingers, palm and thumb which were likely juxtaposed to form conformational epitopes. Molecular docking revealed that the antibodies covered the RdRp catalytic groove. The transbodies await further studies for in vivo role in inhibiting HCV replication.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antiviral Agents/immunology , Hepacivirus/genetics , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Binding Sites , Camelus , Cell Line , Epitopes/immunology , Escherichia coli/genetics , Hepacivirus/immunology , Humans , Molecular Sequence Data , Polymorphism, Restriction Fragment Length , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/immunology , Sequence Alignment , Transfection , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/immunology , Virus Replication
6.
Vaccine ; 28(41): 6765-77, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20688037

ABSTRACT

A non-egg, non-culture based influenza vaccine that intervenes large influenza outbreaks and protects against heterosubtypic infections is needed. Candidates of such vaccine are likely to be conserved influenza virus proteins or their coding DNA. The vaccine must be conveniently produced at reasonable cost, safe, highly immunogenic and should be able to recall rapidly the immunological memory upon the antigenic re-exposure. In this study vaccines made of full length recombinant NP and M2 of the H5N1 influenza A virus were entrapped either alone or together into liposome (L) made of phosphatidylcholine and cholesterol. The vaccines (L-NP, L-M2 or L-NP+M2) and mocks (L or PBS) were safe without causing any adverse reaction in the intramuscularly injected mice. They were readily immunogenic at a single dose and a recalled response could be detected within one day post booster. Cytokine and antibody data indicated that the vaccines induced a Th1 bias immune response. NP containing vaccines stimulated a marked increase of cytotoxic lymphocytes, i.e., CD8(+), intracellular IFNγ(+) cells, while M2 containing vaccines elicited good antibody response which neutralized infectivity of heterologous influenza viruses. Although the three vaccines elicited different immunological defense factors; nevertheless, they similarly and readily abrogated lung histopathology mediated by viruses belonging to different H5N1 clade/subclade and heterosubtypes including swine H1N1 and human H1N1/2009 viruses. They protected the vaccinated mice against lethal challenges with mouse adapted avian H5N1 virus. The liposome adjuvanted vaccines which demonstrated high protective efficacy in mice warrant testing further in a non-rodent model as well as in humans.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Liposomes/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/blood , Antibody Formation , Cross Protection , Cytokines/immunology , Influenza A Virus, H1N1 Subtype/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Neutralization Tests , Nucleocapsid Proteins , Orthomyxoviridae Infections/immunology , RNA-Binding Proteins/immunology , Recombinant Proteins/immunology , Vaccines, Synthetic/immunology , Viral Core Proteins/immunology , Viral Matrix Proteins/immunology
7.
Bioconjug Chem ; 21(7): 1134-41, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20560610

ABSTRACT

A cell penetrating format of human single chain antibody (HuScFv) specific to matrix protein (M1) of influenza A virus was produced by molecular linking of the gene sequence encoding the HuScFv (huscfv) to a protein transduction domain, i.e., penetratin (PEN) of the Drosophila homeodomain. DNA of a recombinant phagemid vector carrying the huscfv was used as a platform template in a three-step PCR for generating a nucleotide sequence encoding a 16 amino acid PEN peptide. The PEN-HuScFv had negligible cytotoxicity on living MDCK cells. They were readily translocated across the cell membrane and bound to native M1 in the A/H5N1-infected cells as revealed by immunofluorescent confocal microscopy. The PEN-HuScFv, when used to treat the influenza virus infected cells, reduced the number of viruses released from the cells. In conclusion, the cell penetrating M1-specific HuScFv, a transbody, produced in this study affected the influenza A virus life cycle in living mammalian cells. While the molecular mechanisms of the PEN-HuScFv need more investigation, the reagent warrants further testing in animals before developing it into a human immunotherapeutic anti-influenza formula.


Subject(s)
Antibody Specificity , Influenza A virus/drug effects , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , Viral Matrix Proteins/immunology , Virus Replication/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell-Penetrating Peptides , Humans , Influenza A virus/growth & development , Influenza A virus/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
8.
J Biol Chem ; 285(13): 9657-9666, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20093370

ABSTRACT

The current treatment of botulism is to administer animal-derived antitoxin, which frequently causes severe adverse reactions in the recipients. In this study, a heavy chain antibody fragment (VH/V(H)H) phage display library was constructed by amplification of the immunoglobulin genes of a nonimmune camel, Camelus dromedarius, using primers specific to human VH gene segments. A recombinant light chain of type A botulinum toxin, BoTxA/LC, with zinc endoprotease activity was used in phage bio-panning to select phage clones displaying BoTxA/LC-bound VH/V(H)H. Soluble VH/V(H)H were produced and purified from 10 VH/V(H)H phagemid-transformed E. coli clones. Complementary determining regions (CDRs) and immunoglobulin frameworks (FRs) of the 10 camel VH/V(H)H-deduced amino acid sequences were determined. FR2 sequences of two clones showed a hallmark of camel V(H)H, i.e. (F/Y)(42)E(49)R(50)(G/F)(52). The remaining eight clones had an FR2 amino acid tetrad of conventional VH, i.e. V(42)G(49)L(50)W(52). V(H)H of one clone (V(H)H17) neutralized the SNAP25 hydrolytic activity of BoTxA/LC, whereas mouse polyclonal anti-BoTxA/LC did not have such activity. Mimotope sequences of V(H)H17 matched with the 194-206 amino acid residues of BoTxA/LC, which are located near the S'1 subsite of the catalytic cleft of the enzyme. Molecular docking revealed that CDR3 of the V(H)H17 bound to epitope in the toxin enzymatic cleft. Therefore, the BoTxA/LC neutralization by the V(H)H17 should be due to the V(H)H insertion into the enzymatic cleft of the toxin, which is usually inaccessible to a conventional antibody molecule. This antibody fragment warrants further development as a therapeutic agent for botulism.


Subject(s)
Antibodies/chemistry , Botulinum Toxins, Type A/metabolism , Metalloproteases/chemistry , Zinc/chemistry , Amino Acid Sequence , Animals , Camelus , Humans , Immunoglobulin Variable Region/chemistry , Male , Mice , Mice, Inbred ICR , Molecular Sequence Data , Peptide Library , Protein Conformation , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid
9.
Antivir Ther ; 14(2): 221-30, 2009.
Article in English | MEDLINE | ID: mdl-19430097

ABSTRACT

BACKGROUND: Human antibodies that interfere with the biological activity of haemagglutinins (HAs) of influenza viruses have high potential as an antiviral agent. METHODS: Human single-chain antibody fragments (HuScFv) to recombinant and native HAs of the influenza virus H5N1 subtype were produced using a human antibody phage display library with the intention to increase the therapeutic arsenal against this highly pathogenic virus. RESULTS: The HuScFv inhibited HA activity and neutralized infectivity of both homologous and heterologous strains and clades of the H5N1 subtype in Madin-Darby canine kidney cell cultures. Intraperitoneally injected HuScFv also mediated immunotherapeutic protection in mice that were intranasally challenged with highly pathogenic H5N1 viruses belonging to different strains and clades. CONCLUSIONS: Our data indicate that it might be worth pursuing these HuScFv further for future consideration as candidates for influenza intervention and treatment.


Subject(s)
Antibodies, Viral/administration & dosage , Influenza A Virus, H5N1 Subtype , Influenza, Human/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Cell Line , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunization, Passive , Immunoglobulin Fragments/administration & dosage , Immunologic Factors/administration & dosage , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/immunology , Injections, Intraperitoneal , Mice
10.
J Virol Methods ; 159(1): 105-11, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19442853

ABSTRACT

Matrix protein (M1) is predominant and has pivotal role in the influenza A virus replication and assembly. It is therefore an attractive target for antiviral drugs, siRNA studies, and therapeutic antibodies. Nevertheless, therapeutic antibody that interferes with the M1 multiplex function has never been developed. In this study, human single monoclonal antibody fragments (HuScFvs) to M1 were generated. Full length recombinant M1 (rM1) was produced from cDNA prepared from genome of highly pathogenic avian influenza virus, A/H5N1. The rM1 was used as an antigen in phage bio-panning to select phage clones displaying HuScFv from a human antibody phage display library. Several phage clones displaying HuScFv bound to the rM1 and harboring the respective huscfv gene inserts were isolated. RFLP experiments revealed multiple DNA banding patterns which indicated epitope/affinity diversity of the HuScFv. The HuScFv were tested for their binding to native M1 of homologous and heterologous influenza A viruses using ELISA as well as incorporating immunostaining and immunofluorescence studies with infected MDCK cells. One such protein produced from a selected phage clone blocked binding of M1 to viral RNA. The HuScFv in their in vivo functional format, e.g. cell-penetrating molecules, should be developed and tested as a broad spectrum anti-A/influenza.


Subject(s)
Antibodies, Monoclonal/immunology , Influenza A virus/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/immunology , Antibody Specificity , Dogs , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Subunits/immunology , In Situ Hybridization, Fluorescence , Influenza A virus/metabolism , Mice , Peptide Library , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Viral Matrix Proteins/biosynthesis
11.
Protein Eng Des Sel ; 22(5): 305-12, 2009 May.
Article in English | MEDLINE | ID: mdl-19297536

ABSTRACT

Patients with leptospirosis are commonly treated with antibiotics. Jarisch-Herxheimer reaction caused by toxic bacterial substances massively released as a result of the antibiotic mediated-bacterial lysis occurs in some patients which may aggravate the existing severe clinical manifestations. In this study, a humanized-murine single-chain monoclonal antibody (HuScFv) was produced and tested as an alternative of antibiotics for treatment of leptospirosis. Complementary DNA was prepared from total RNA of a murine hybridoma clone secreting monoclonal antibody (MAb) specific to LipL32 of pathogenic Leptospira spp. The MAb had therapeutic efficacy in Leptospira challenged hamsters. The VH and VL coding sequences were amplified using the cDNA as a template. The sequences were linked to form a single-chain variable murine DNA fragment (muscFv). CDR sequences of the muscFv were grafted onto the best matching human VH and VL immunoglobulin frameworks. After cloning of the humanized murine DNA sequences (huscFv) into a phagemid vector and the vector was introduced into competent Escherichia coli, the HuScFv was produced. On the same weight basis, the HuScFv possessed equal neutralizing activities to the murine ScFv counterpart against heterologous Leptospira-mediated hemolysis in vitro and rescued hamsters from a heterologous Leptospira lethal challenge. The HuScFv antibody has high therapeutic potential as an alternative to antibiotics for human leptospirosis, especially for drug hypersensitive patients.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotherapy/methods , Leptospira/immunology , Leptospirosis/therapy , Animals , Antibodies, Monoclonal/genetics , Bacterial Outer Membrane Proteins/immunology , Blotting, Western , Cloning, Molecular , Computational Biology , Cricetinae , DNA Primers/genetics , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Genetic Vectors/genetics , Humans , Leptospirosis/immunology , Lipoproteins/immunology , Mice , Neutralization Tests
12.
J Proteomics ; 72(2): 270-82, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19162253

ABSTRACT

Animal derived anti-Naja. kaouthia (Thai cobra) venom is used for specific treatment of the snake bitten victims. Many recipients develop allergic reaction or anti-isotype response which causes serum sickness. A better therapeutic antibody is needed. In this study, long alpha-neurotoxin was purified from the N. kaouthia holovenom and verified by 2D-LC/MS-MS. The toxin was used as antigen in a phage bio-panning to select phage clones displaying human single chain variable antibody fragments (HuScFv) from a phage display antibody library constructed from immunoglobulin genes of non-immunized Thai blood donors. HuScFv that specifically bound to the neurotoxin were produced from huscfv-phagemid transformed E. coli clones and affinity purified. The HuScFv could neutralize toxicity of the N. kaouthia neurotoxin and rescued the envenomized mice from the neurotoxin mediated lethality. Peptide mimotope of the neutralizing HuScFv matched with an amino acid sequence (epitope) located in the loop-3 of the N. kaouthia long alpha-neurotoxin which functions in acetylcholine receptor binding. The mimotope is also similar to peptide sequences found on other snake venom neurotoxins implying a possibility of the HuScFv to exert pan-neutralizing activity against multiple snake neurotoxins.


Subject(s)
Antibodies, Monoclonal/chemistry , Elapid Venoms/analysis , Immunoglobulin Fragments/analysis , Neurotoxins/analysis , Amino Acid Sequence , Animals , Base Sequence , Chromatography, Liquid/methods , Elapid Venoms/chemistry , Elapidae , Immunoglobulin Fragments/chemistry , Male , Mice , Mice, Inbred ICR , Molecular Sequence Data , Neurotoxins/chemistry , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Hybridoma (Larchmt) ; 27(6): 453-65, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19108618

ABSTRACT

A non-culture-based leptospirosis vaccine that cross-protects against infection caused by heterologous Leptospira spp. should replace the currently available products, which are qualitatively and quantitatively inadequate. With that in mind, two murine hybridomas secreting monoclonal antibodies (MAb) binding only to homogenates of pathogenic Leptospira spp., and not of the saprophytic L. biflexa, serogroup Patoc, serovar Patoc, were produced. The MAbs of both clones neutralized Leptospira-mediated human red blood cell lysis in vitro and rescued hamsters from lethal infection with heterologous Leptospira spp. The orthologous Leptospira spp. protein carrying the MAb epitope(s) was identified by two-dimensional gel electrophoresis (2DE)-based proteomics and database search. The epitopes of the MAbs were located on the major outer membrane protein LipL32 of the pathogenic Leptospira spp. The MAbs in their humanized version are potential leptospirosis immunotherapeutics. They are also suitable as detection reagents in antigen-based assays for the rapid diagnosis of leptospirosis. Recombinant LipL32 is a good candidate for a broad spectrum, non-culture-based leptospirosis vaccine.


Subject(s)
Antibodies, Monoclonal/chemistry , Bacterial Outer Membrane Proteins/chemistry , Hybridomas/metabolism , Leptospira/metabolism , Leptospirosis/prevention & control , Lipoproteins/chemistry , Animals , Chromatography, Liquid/methods , Cricetinae , Epitopes/chemistry , Erythrocytes/metabolism , Hemolysis , Mass Spectrometry/methods , Mesocricetus , Mice , Proteomics/methods
14.
Asian Pac J Allergy Immunol ; 26(1): 23-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18595527

ABSTRACT

Current anti-influenza drugs target the viral neuraminidase or inhibit the function of the ion channel M2 protein. Not only is the supply of these drugs unlikely to meet the demand during a large influenza epidemic/ pandemic, but also has an emergence of drug resistant influenza virus variants been documented. Thus a new effective drug or antiviral alternative is required. The influenza virus RNA polymerase complex consists of nucleoproteins (NP) that bind to three polymerase subunits: two basic polymerases, PB1 and PB2, and an acidic polymerase (PA). These proteins play a pivotal role in the virus life cycle; thus they are potential targets for the development of new anti-influenza agents. In this study, we produced human monoclonal antibodies that bound to the influenza A polymerase proteins by using a human antibody phage display library. Complementary DNA was prepared from the total RNA of a highly pathogenic avian influenza (HPAI) virus: A/duck/Thailand/144/2005(H5N1). The cDNA synthesized from the total virus RNA was used as template for the amplification of the gene segments encoding the N-terminal halves of the PB1, PB2 and PA polymerase proteins which encompassed the biologically active portions of the respective proteins. The cDNA amplicons were individually cloned into appropriate vectors and the recombinant vectors were introduced into Escherichia coli bacteria. Transformed E. coli clones were selected, and induced to express the recombinant proteins. Individually purified proteins were used as antigens in bio-panning to select the phage clones displaying specific human monoclonal single chain variable fragments (HuScFv) from a human antibody phage display library constructed from Thai blood donors in our laboratory. The purified HuScFv that bound specifically to the recombinant polymerase proteins were prepared. The inhibitory effects on the biological functions of the respective polymerase proteins should be tested. We envisage the use of the HuScFv in their cell penetrating version (transbodies) as an alternative influenza therapeutic to current anti-virus drugs.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Variable Region/immunology , Influenza A Virus, H5N1 Subtype/immunology , RNA-Dependent RNA Polymerase/immunology , Viral Proteins/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Specificity , Cloning, Molecular , Genetic Vectors , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Influenza A Virus, H5N1 Subtype/enzymology , Peptide Library , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
15.
Asian Pac J Allergy Immunol ; 25(1): 53-73, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17891922

ABSTRACT

In this study, proteomes of two pathogenic Leptospira spp., namely L. interrogans, serogroup Icterohaemorrhagiae, serovar Copenhageni and L. borgpetersenii, serogroup Tarassovi, serovar Tarassovi, were revealed by using two dimensional gel electrophoresis (2DE)-based-proteomics. Bacterial cells were disrupted in a lysis buffer containing 30 mM Tris, 2 M thiourea, 7 M urea, 4% CHAPS, 2% IPG buffer pH 3-10 and protease inhibitors and then subjected to sonication in order to solubilize as much as possible the bacterial proteins. The 2DE-separated components of both Leptospira homogenates were blotted individually onto membranes and antigenic components (immunomes) were revealed by probing the blots with immune serum of a mouse readily immunized with the homogenate of L. interrogans, serogroup Icterohaemorrhagiae, serovar Copenhageni. The immunogenic proteins of the two pathogenic Leptospira spp. could be grouped into 10 groups. These are: 1) proteins involved in the bacterial transcription and translation including beta subunit transcription anti-termination protein of DNA polymerase III, elongation factors Tu and Ts, and tRNA (guanine-N1)-methyltransferase; 2) proteins functioning as enzymes for metabolisms and nutrient acquisition including acetyl-Co-A acetyltransferase, putative glutamine synthetase, glyceraldehyde-3-phospahte dehydrogenase, NifU-like protein, 3-oxoacyl-(acyl-carrier-protein) reductase, oxidoreductase, sphingomyelinase C precursor, spermidine synthase, beta subunit of succinyl-CoA synthetase, and succinate dehydrogenase iron-sulfur subunit; 3) proteins/enzymes necessary for energy and electron transfer, i.e. electron transfer flavoprotein, and proton-translocating transhydrogenase; 4) enzymes for degradation of misfolded proteins, i.e. ATP-dependent Clp protease; 5) molecular chaperone, i.e. 60 kDa chaperonin; 6) signal transduction system, i.e. response regulator; 7) protein involved in immune evasion in host, i.e. peroxiredoxin; 8) cell structure proteins including MreB (cytoskeletal) and flagellin/ periplasmic flagellin; 9) lipoproteins/outer membrane proteins: LipL32, LipL41, LipL45 and OmpL1; and 10) various hypothetical proteins. Many immunogenic proteins are common to both Leptospira spp. These proteins not only are the diagnostic targets but also have potential as candidates of a broad spectrum leptospirosis vaccine especially the surface exposed components which should be vulnerable to the host immune effector factors.


Subject(s)
Antigens, Bacterial/immunology , Leptospira interrogans serovar icterohaemorrhagiae/immunology , Leptospira/immunology , Proteome/immunology , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/isolation & purification , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Leptospira/chemistry , Leptospira/isolation & purification , Leptospira interrogans serovar icterohaemorrhagiae/chemistry , Leptospirosis/microbiology , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Proteomics
16.
Asian Pac J Allergy Immunol ; 25(1): 75-82, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17891923

ABSTRACT

Available leptospirosis vaccines made up of inactivated bacteria or their membrane components elicit immunity which is serovar specific and unsatisfactory immunological memory. A vaccine that protects across Leptospira serogroups/serovars, i.e. broad spectrum, and induces long-lasting memory is needed for both human and veterinary uses. In this study, a plasmid DNA vaccine was constructed from cloning gene encoding a transmembrane porin protein, OmpL1, of pathogenic Leptospira interrogans, serogroup Icterohaemorrhagiae, serovar Copenhageni into a mammalian expression vector pcDNA3.1(+). The protective efficacy of the ompL1-pcDNA3.1(+) plasmid DNA vaccine was studied by immunizing hamsters intramuscularly with three doses of the vaccine (100 microg per dose) at two week intervals. The empty pcDNA3.1(+) and PBS were used as mock as negative vaccine controls, respectively. All animals were challenged with the heterologous Leptospira interrogans, serogroup Pomona, serovar Pomona (10 LD50), at one week after the last vaccine booster. The ompL1-pcDNA3.1(+) plasmid DNA vaccine rescued some vaccinated animals from the lethal challenge and delayed death time, reduced morbidity, e.g. fever, and/or the numbers of Leptospira in the tissues of the vaccinated animals. While the results are encouraging, further studies are needed to optimize the immunization schedule, vaccine dosage and formulation in order to maximize the efficacy of the vaccine.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Leptospira interrogans serovar icterohaemorrhagiae/immunology , Leptospirosis/prevention & control , Vaccines, DNA/immunology , Animals , Bacterial Vaccines/administration & dosage , COS Cells , Chlorocebus aethiops , Cricetinae , Cross Reactions , Female , Leptospira interrogans serovar icterohaemorrhagiae/classification , Leptospirosis/immunology , Leptospirosis/microbiology , Mesocricetus , Plasmids , Vaccines, DNA/administration & dosage
17.
Asian Pac J Allergy Immunol ; 25(4): 225-32, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18402296

ABSTRACT

Existing serological methods for diagnosis of leptospirosis are still unsatisfactorily due mainly to their low accuracy. In this study, serum samples of 18 clinically diagnosed-, IgM dipstick positive-, MAT positive-leptospirosis patients (group 1) were analyzed by IgG Western blotting against SDS-PAGE separated-whole cell homogenates of pathogenic and non-pathogenic Leptospira spp. belonging to 20 serovars of 15 serogroups. The samples of group 1 were collected from the patients at days 3 to 10 after the fever onset (fist samples). Second and third samples could be obtained from 4 patients. Sera of the 22 patients with other febrile illnesses (group 2) and 22 healthy counterparts (group 3) were used as patient- and normal- controls, respectively. Irrespective of the serovar or serogroup of the pathogenic Leptospira spp. used as antigen in the Western blotting, all of the 18 sera of patients with leptospirosis (group 1) gave characteristic diffuse antigen-antibody reactive bands located at approximately 35-38 and 22-26 kDa; and thus 100% diagnostic sensitivity of the Western blot assay. Some serum samples of the leptospirosis patients also reacted to components located at 80-100, approximately 70, 60, 54, and 48 kDa. More bands or the early recognized bands with increased intensity were observed when tested the second and third samples. The characteristic bands were not seen when homogenates of L. biflexa, serogroup Semaranga, serovar Patoc (saprophytic) and L. biflexa, serogroup Andamana, serovar Andamana (non-pathogenic but can infect host) were used in the assay. Sera of groups 2 and 3 did not react to the components at the seven locations implying 100% diagnostic specificity of the IgG Western blot assay. While awaiting validation with more patients' samples, the IgG Western Blot analysis aiming at the detection of the characteristic antigen-antibody reactive bands described in this study has high potential for early, rapid, simple and accurate diagnosis of human leptospirosis.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Immunoglobulin G/immunology , Leptospira/immunology , Leptospirosis/diagnosis , Leptospirosis/immunology , Antibodies, Bacterial/blood , Antibody Specificity/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunoglobulin M/immunology , Leptospira/pathogenicity , Leptospirosis/blood , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...