Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Sci Rep ; 11(1): 12987, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34155267

ABSTRACT

Dengue virus (DENV), from the genus flavivirus of the family flaviviridae, causes serious health problems globally. Human monoclonal antibodies (HuMAb) can be used to elucidate the mechanisms of neutralization and antibody-dependent enhancement (ADE) of DENV infections, leading to the development of a vaccine or therapeutic antibodies. Here, we generated eight HuMAb clones from an Indonesian patient infected with DENV. These HuMAbs exhibited the typical characteristics of weak neutralizing antibodies including high cross-reactivity with other flaviviruses and targeting of the fusion loop epitope (FLE). However, one of the HuMAbs, 3G9, exhibited strong neutralization (NT50 < 0.1 µg/ml) and possessed a high somatic hyper-mutation rate of the variable region, indicating affinity-maturation. Administration of this antibody significantly prolonged the survival of interferon-α/ß/γ receptor knockout C57BL/6 mice after a lethal DENV challenge. Additionally, Fc-modified 3G9 that had lost their in vitro ADE activity showed enhanced therapeutic potency in vivo and competed strongly with an ADE-prone antibody in vitro. Taken together, the affinity-matured FLE-targeting antibody 3G9 exhibits promising features for therapeutic application including a low NT50 value, potential for treatment of various kinds of mosquito-borne flavivirus infection, and suppression of ADE. This study demonstrates the therapeutic potency of affinity-matured FLE-targeting antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antigens, Viral , Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/virology , Epitopes , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Affinity/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Antiviral Agents/therapeutic use , Dengue/drug therapy , Dengue/immunology , Dengue Virus/immunology , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Humans , Hybridomas , Mice , Models, Molecular , Neutralization Tests , Protein Conformation , Recombinant Proteins , Structure-Activity Relationship
2.
Sci Rep ; 10(1): 21561, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33299049

ABSTRACT

In a secondary dengue virus (DENV) infection, the presence of non-neutralizing antibodies (Abs), developed during a previous infection with a different DENV serotype, is thought to worsen clinical outcomes by enhancing viral production. This phenomenon is called antibody-dependent enhancement (ADE) of infection, and it has delayed the development of therapeutic Abs and vaccines against DENV, as they must be evaluated for the potential to induce ADE. Unfortunately, limited replication of DENV clinical isolates in vitro and in experimental animals hinders this evaluation process. We have, therefore, constructed a recombinant chimeric flavivirus (DV2ChimV), which carries premembrane (prM) and envelope (E) genes of type 2 DENV (DENV-2) R05-624 clinical (Thai) isolate in a backbone of Japanese encephalitis virus (Nakayama strain). DENV E-protein is the most important viral target, not only for neutralizing Abs, but also for infection-enhancing Abs. In contrast to DENV-2 R05-624, DV2ChimV replicated efficiently in cultured mammalian cells and was lethal in interferon-α/ß-γ-receptor double-knockout mice. With DV2ChimV, we were able to perform neutralization assays, in vitro and in vivo ADE assays, and in vivo protection assays. These results suggest that the chimeric virus is a powerful tool for evaluation of Abs against DENV.


Subject(s)
Antibodies, Viral/immunology , Dengue Virus/metabolism , Dengue/immunology , Flavivirus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Dengue/virology , Dengue Virus/genetics , Mice , Vero Cells , Viral Envelope/immunology , Viral Envelope Proteins/genetics
3.
Immunobiology ; 223(3): 319-326, 2018 03.
Article in English | MEDLINE | ID: mdl-29107382

ABSTRACT

Influenza virus causes acute respiratory infection in humans, and is a major public health concern globally. Antibodies play a central role in host protection against influenza virus. We isolated human monoclonal antibodies (hMAb) 206-2-4 and 201-6-8 by a human hybridoma protocol that neutralized various but distinct influenza virus (IFV) A/H1N1 strains, including 2009 pandemic strains. The half-inhibitory concentration of 206-2-4 and 201-6-8 against A/H1N1pdm09 strains was 2-100ng/mL and 5-20µg/mL, respectively. Prophylactic and therapeutic potencies of 206-2-4 were demonstrated in a mouse model of IFV infection at i.p. dosages of 0.25 and 2.5mg/kg, respectively, suggesting that 206-2-4 is one of the most potent hnMAbs against IFV reported thus far. The Ig genes of 206-2-4 and 201-6-8 were originated from distinct germ line repertoires, and accompanied by 63 and 23 somatic hypermutations, respectively. The hemagglutination inhibitory activity indicated that the mechanism of neutralization was to interfere the virus-receptor interaction. The binding epitope of the two antibodies was mapped to hemagglutinin 1 (HA1) amino acid residues 111-120. Additional interaction between the antibody and the HA1 globular head was necessary for neutralization. Such hnMAbs bearing distinct binding epitope have been rarely reported. The potency is likely due to the coverage of a wide surface area of HA protein by these hnMABs. IFV is a highly variable. Our knowledge on the mechanisms by which these cross-reactive hnMAbs function should help design a novel immunogen for the development of a vaccine effective against broader spectrum of IFV strains.


Subject(s)
Genes, Immunoglobulin , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Cells, Cultured , Cross Reactions , Dogs , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Hybridomas , Influenza, Human/epidemiology , Japan/epidemiology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Pandemics
4.
Biologicals ; 44(5): 394-402, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27464991

ABSTRACT

In previous studies, hybridomas producing human immunoglobulin G, the antibodies 5E4 and 5A7 against influenza A and B virus were established using a novel human lymphocyte fusion partner, SPYMEG. In the present study, we succeeded in achieving the recombinant production and secretion of 5E4 and 5A7 in Chinese hamster ovary cells. Our N-glycan analysis by intact-mass detection and liquid chromatography mass spectrometry showed that recombinant 5E4 and 5A7 have one N-glycan and the typical mammalian-type N-glycan structures similar to those in hybridomas. However, the glycan distribution was slightly different among these antibodies. The amount of high-mannose-type structures was under 10% of the total N-glycans of recombinant 5E4 and 5A7, compared to 20% of the 5E4 and 5A7 produced in hybridomas. The amount of galactosylated N-glycans was increased in recombinants. Approximately 80% of the N-glycans of all antibodies was fucosylated, and no sialylated N-glycan was found. Recombinant 5E4 and 5A7 neutralized pandemic influenza A virus specifically, and influenza B virus broadly, quite similar to the 5E4 and 5A7 produced in hybridomas, respectively. Here we demonstrated that recombinants of antibodies identified from hybridomas fused with SPYMEG have normal N-glycans and that their neutralizing activities bear comparison with those of the original antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunoglobulin G/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/blood , Cell Fusion/methods , Cricetinae , Humans , Hybridomas/immunology , Hybridomas/metabolism , Immunoglobulin G/biosynthesis , Lymphocytes/immunology , Lymphocytes/metabolism
5.
Monoclon Antib Immunodiagn Immunother ; 35(2): 73-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26974561

ABSTRACT

Heparin-binding EGF-like growth factor (HB-EGF) is a member of the EGF family of growth factors that bind to and activate the EGF receptor (EGFR/ErbB1) and ErbB4. HB-EGF plays pivotal roles in pathophysiological processes, including cancer. Thus, monoclonal antibodies (mAbs) for HB-EGF detection could be an important tool in the therapeutic diagnosis of HB-EGF-related cancers and other diseases. However, few mAbs, especially those applicable for immunohistochemistry (IHC), have been established to date. In this study, we generated a clone of hybridoma-derived mAb 2-108 by immunizing mice with recombinant human HB-EGF protein expressed by human cells. The mAb 2-108 specifically bound to human HB-EGF but not to mouse HB-EGF and was successful in immunoblotting, even under reducing conditions, immunoprecipitation, and immunofluorescence for unfixed as well as paraformaldehyde-fixed cells. Notably, this mAb was effective in IHC of paraffin-embedded tumor specimens. Epitope mapping analysis showed that mAb 2-108 recognized the N-terminal prodomain in HB-EGF. These results indicate that this new anti-HB-EGF mAb 2-108 would be useful in the diagnosis of HB-EGF-related cancers and would be a strong tool in both basic and clinical research on HB-EGF.


Subject(s)
Antibodies, Monoclonal/immunology , Epidermal Growth Factor/immunology , Heparin-binding EGF-like Growth Factor/immunology , Neoplasms/immunology , Animals , Cell Line, Tumor , Epidermal Growth Factor/isolation & purification , Epitope Mapping , ErbB Receptors/immunology , ErbB Receptors/isolation & purification , Heparin-binding EGF-like Growth Factor/isolation & purification , Humans , Immunohistochemistry , Mice , Neoplasms/diagnosis , Paraffin
6.
Antiviral Res ; 124: 61-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26522769

ABSTRACT

Antibody-dependent enhancement (ADE) of dengue virus (DENV) infectivity is thought to play a crucial role in severe dengue disease. It occurs when pre-existing sub-neutralizing anti-DENV antibody (Ab) produced from a primary infection encounters a DENV serotype different from that of the initial infection and forms immune complexes, which enable the efficient infection of Fcγ receptor-bearing cells. However, the exact role played by Abs during a secondary infection of patients remains unknown. We previously obtained a broadly cross-reactive neutralizing IgG1 human monoclonal anti-DENV envelope (E) Ab (HuMAb) D23-1G7C2-IgG1 from a DENV-infected patient; however, D23-1G7C2-IgG1 had ADE activity. With the aim of being able to reduce the ADE activity, we exchanged the Fc region of D23-1G7C2 to generate Abs bearing each of the three other IgG subclasses (IgG2-4). In addition, N297A, a mutation known to reduce the affinity of the IgG1 Fc region for Fcγ receptors, was introduced into D23-1G7C2-IgG1. Swapping D23-1G7C2-IgG1 to IgG2 or IgG4 subclasses reduced ADE activity in FcγRI and FcγRII-bearing THP-1 cells. By contrast, in FcγRII-bearing K562 cells, the change to IgG2 increased ADE activity. Introducing the N297A mutation into D23-1G7C2-IgG1 resulted in a marked reduction in ADE activity in both cell types. Compared to D23-1G7C2-IgG1, D23-1G7C2-IgG1-N297A was less protective in IFN-α/ß/γ receptor knockout mice infected with a lethal dose of recombinant chimeric DENV, carrying prME of DENV-2 in Japanese encephalitis virus (80% vs. 40% survival, respectively). These observations provide valuable information regarding the use of recombinant Abs as therapeutics.


Subject(s)
Antibodies, Viral/pharmacology , Dengue Virus/drug effects , Dengue/therapy , Dengue/virology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Antibody-Dependent Enhancement/drug effects , Cross Reactions , Dengue Virus/immunology , HEK293 Cells , Humans , Immunoglobulin G/immunology , K562 Cells , Mice , Mutation , Protein Engineering , Receptors, IgG/genetics , Severe Dengue/immunology , Vero Cells , Viral Envelope Proteins/immunology
7.
Biochem Biophys Res Commun ; 452(3): 865-70, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25204499

ABSTRACT

Most neutralizing antibodies elicited during influenza virus infection or by vaccination have a narrow spectrum because they usually target variable epitopes in the globular head region of hemagglutinin (HA). In this study, we describe a human monoclonal antibody (HuMAb), 5D7, that was prepared from the peripheral blood lymphocytes of a vaccinated volunteer using the fusion method. The HuMAb heterosubtypically neutralizes group 1 influenza A viruses, including seasonal H1N1, 2009 pandemic H1N1 (H1N1pdm) and avian H9N2, with a strong hemagglutinin inhibition activity. Selection of an escape mutant showed that the HuMAb targets a novel conformational epitope that is located in the HA head region but is distinct from the receptor binding site. Furthermore, Phe114Ile substitution in the epitope made the HA unrecognizable by the HuMAb. Amino acid residues in the predicted epitope region are also highly conserved in the HAs of H1N1 and H9N2. The HuMAb reported here may be a potential candidate for the development of therapeutic/prophylactic antibodies against H1 and H9 influenza viruses.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Cross Protection , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Influenza, Human/prevention & control , Amino Acid Sequence , Animals , Antigens, Viral/chemistry , Antigens, Viral/immunology , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Hybridomas/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Influenza, Human/virology , Male , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , Vaccination , Young Adult
8.
Biochem Biophys Res Commun ; 450(1): 42-8, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24858683

ABSTRACT

Influenza viruses are a continuous threat to human public health because of their ability to evolve rapidly through genetic drift and reassortment. Three human monoclonal antibodies (HuMAbs) were generated in this study, 1H11, 2H5 and 5G2, and they cross-neutralize a diverse range of group 1 influenza A viruses, including seasonal H1N1, 2009 pandemic H1N1 (H1N1pdm) and avian H5N1 and H9N2. The three HuMAbs were prepared by fusing peripheral blood lymphocytes from an H1N1pdm-infected patient with a newly developed fusion partner cell line, SPYMEG. All the HuMAbs had little hemagglutination inhibition activity but had strong membrane-fusion inhibition activity against influenza viruses. A protease digestion assay showed the HuMAbs targeted commonly a short α-helix region in the stalk of the hemagglutinin. Furthermore, Ile45Phe and Glu47Gly double substitutions in the α-helix region made the HA unrecognizable by the HuMAbs. These two amino acid residues are highly conserved in the HAs of H1N1, H5N1 and H9N2 viruses. The HuMAbs reported here may be potential candidates for the development of therapeutic antibodies against group 1 influenza viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Hemagglutinins/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/virology , Animals , Antibodies, Viral/immunology , Cell Line , Haplorhini , Humans , Kidney/immunology , Kidney/virology , Neutralization Tests
9.
Biologics ; 7: 175-87, 2013.
Article in English | MEDLINE | ID: mdl-23983454

ABSTRACT

BACKGROUND: Hybridomas that produce human monoclonal antibodies (HuMAbs) against Dengue virus (DV) had been prepared previously using peripheral blood lymphocytes from patients with DV during the acute and convalescent phases of a secondary infection. Anti-DV envelope glycoprotein (E) 99 clones, anti-DV premembrane protein (prM) 8 clones, and anti-DV nonstructural protein 1 (NS1) 4 clones were derived from four acute-phase patients, and anti-DV E 2 clones, anti-DV prM 2 clones, and anti-DV NS1 8 clones were derived from five convalescent-phase patients. METHODS AND RESULTS: In the present study, we examined whether these clones cross-reacted with Japanese encephalitis virus (JEV), which belongs to the same virus family. Forty-six of the above-described 99 (46/99) anti-E, 0/8 anti-prM, and 2/4 anti-NS1 HuMAbs from acute-phase, and 0/2 anti-E, 0/2 anti-prM, and 5/8 anti-NS1 HuMAbs from convalescent-phase showed neutralizing activity against JEV. Thus, most of the anti-E and anti-NS1 (but not the anti-prM) antibodies cross-reacted with JEV and neutralized this virus. Interestingly, 3/46 anti-E HuMAbs derived from acute-phase patients and 3/5 anti-NS1 HuMAbs from convalescent-phase patients showed particularly high neutralizing activity against JEV. Consequently, the HuMAbs showing neutralization against JEV mostly consisted of two populations: one was HuMAbs recognizing DV E and showing neutralization activity against all four DV serotypes (complex-type) and the other was HuMAbs recognizing DV NS1 and showing subcomplex-type cross-reaction with DV. CONCLUSION: Anti-DV E from acute phase (46/99) and anti-DV NS1 (7/12) indicate neutralizing activity against JEV. In particular, three of 46 anti-DV E clones from acute phase and three of five anti-NS1 clones from convalescent phase showed strong neutralizing activity against JEV.

10.
Immunity ; 38(5): 1050-62, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23602766

ABSTRACT

Cord factor, also called trehalose-6,6'-dimycolate (TDM), is a potent mycobacterial adjuvant. We herein report that the C-type lectin MCL (also called Clec4d) is a TDM receptor that is likely to arise from gene duplication of Mincle (also called Clec4e). Mincle is known to be an inducible receptor recognizing TDM, whereas MCL was constitutively expressed in myeloid cells. To examine the contribution of MCL in response to TDM adjuvant, we generated MCL-deficient mice. TDM promoted innate immune responses, such as granuloma formation, which was severely impaired in MCL-deficient mice. TDM-induced acquired immune responses, such as experimental autoimmune encephalomyelitis (EAE), was almost completely dependent on MCL, but not Mincle. Furthermore, by generating Clec4e(gfp) reporter mice, we found that MCL was also crucial for driving Mincle induction upon TDM stimulation. These results suggest that MCL is an FcRγ-coupled activating receptor that mediates the adjuvanticity of TDM.


Subject(s)
Cord Factors/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lectins, C-Type/immunology , Membrane Proteins/metabolism , Receptors, IgG/immunology , Adjuvants, Immunologic , Animals , Encephalomyelitis, Autoimmune, Experimental/microbiology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium Infections/immunology , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology
11.
Antiviral Res ; 98(3): 423-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545366

ABSTRACT

Public health concern about dengue diseases, caused by mosquito-borne infections with four serotypes of dengue virus (DENV-1-DENV-4), is escalating in tropical and subtropical countries. Most of the severe dengue cases occur in patients experiencing a secondary infection with a serotype that is different from the first infection. This is believed to be due to antibody-dependent enhancement (ADE), by which one DENV serotype uses pre-existing anti-DENV antibodies elicited in the primary infection to facilitate entry of a different DENV serotype into the Fc receptor-positive macrophages. Recently, we prepared a number of hybridomas producing human monoclonal antibodies (HuMAbs) by using peripheral blood lymphocytes from Thai patients at acute phase of secondary infection with DENV-2. Here, we characterized 17 HuMAbs prepared from two patients with dengue fever (DF) and one patient with dengue hemorrhagic fever (DHF) that were selected as antibodies recognizing viral envelope protein and showing higher neutralization activity to all serotypes. In vivo evaluation using suckling mice revealed near perfect activity to prevent mouse lethality following intracerebral DENV-2 inoculation. In a THP-1 cell assay, these HuMAbs showed ADE activities against DENV-2 at similar levels between HuMAbs derived from DF and DHF patients. However, the F(ab')2 fragment of the HuMAb showed a similar virus neutralization activity as original, with no ADE activity. Thus, these HuMAbs could be one of the therapeutic candidates against DENV infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody-Dependent Enhancement , Dengue Virus/immunology , Dengue/therapy , Adult , Animals , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Coinfection/immunology , Coinfection/virology , Dengue/immunology , Dengue Virus/pathogenicity , Drug Evaluation, Preclinical , Female , Humans , Hybridomas/immunology , Hybridomas/virology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Male , Mice , Mice, Inbred BALB C , Neutralization Tests , Severity of Illness Index , Viral Envelope Proteins/immunology , Virus Internalization , Young Adult
12.
J Biol Chem ; 288(7): 4981-90, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23264630

ABSTRACT

The development of vaccination methods that can overcome the emergence of new types of influenza strains caused by escape mutations is desirable to avoid future pandemics. Here, a novel type of immunogen was designed that targeted the conformation of a highly conserved region of influenza A virus hemagglutinin (HA) composed of two separate sequences that associate to form an anti-parallel ß-sheet structure. Our previous study identified this ß-sheet region as the structural core in the epitope of a characteristic antibody (B-1) that strongly neutralizes a wide variety of strains within the H3N2 serotype, and therefore this ß-sheet region was considered a good target to induce broadly reactive immunity against the influenza A virus. To design the immunogen, residues derived from the B-1 epitope were introduced directly onto a part of enhanced green fluorescent protein (EGFP), whose surface is mostly composed of ß-sheets. Through site-directed mutagenesis, several modified EGFPs with an epitope-mimicking structure embedded in their surface were prepared. Two EGFP variants, differing from wild-type (parental) EGFP by only five and nine residues, induced mice to produce antibodies that specifically bind to H3-type HA and neutralize H3N2 virus. Moreover, three of five mice immunized with each of these EGFP variants followed by a booster with equivalent mCherry variants acquired anti-viral immunity against challenge with H3N2 virus at a lethal dosage. In contrast to conventional methods, such as split HA vaccine, preparation of this type of immunogen requires less time and is therefore expected to be quickly responsive to newly emerged influenza viral strains.


Subject(s)
Epitopes/chemistry , Green Fluorescent Proteins/metabolism , Hemagglutinins/chemistry , Animals , Circular Dichroism , Escherichia coli/metabolism , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hybridomas/metabolism , Immunization , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Mutagenesis , Neutralization Tests , Orthomyxoviridae/immunology , Protein Engineering/methods , Protein Structure, Secondary , Viral Proteins/chemistry
13.
Article in English | MEDLINE | ID: mdl-24437318

ABSTRACT

We conducted this study to determine the clinical variables associated with the production of human immunodeficiency virus type 1 (HIV-1) circulating recombinant form (CRF) 01_AE neutralizing human monoclonal antibodies (NhMAbs) using a hybridoma technique. This cross sectional study was performed in 20 asymptomatic HIV-1-infected Thais. Peripheral blood mononuclear cells (PBMCs) were obtained from each study participant and fused with SPYMEG cells. Culture supernatant collected from growing hybridomas was tested for neutralizing activity against HIV-1 CRF01_AE Env-recombinant viruses. Fifty hybridomas expressing anti-HIV-1 NhMAbs with strong neutralizing activity against at least 1 CRF01_AE Env-recombinant virus were found. A positive association between the numbers of hybridomas produced and the CD4 counts of study participants (p = 0.019) was observed. NhMAb-producing hybridomas with strong neutralizing activity were mostly found in participans diagnosed with HIV-1 infection within the previous 1 year. The HIV-1 viral load was not significantly correlated with the numbers of either established hybridomas or clones expressing anti-HIV-1 NhMAbs with strong neutralizing activity. To our knowledge, this is the first study of NhMAb-producing hybridomas obtained from HIV-1 CRF01_AE-infected populations identified by antibody binding to HIV-1 V3 loop peptide enzyme-linked immunosorbent assay (ELISA) or TRUGENE HIV-1 Genotyping Assay (HIV-1 pol sequence). It provides important criterion to slect study participants with high CD4 counts who produce large numbers of hybridoma clones. The results are valuable for further studies related to nurtalizing antibodies production and HIV-1 vaccine development.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Adult , CD4 Lymphocyte Count , Enzyme-Linked Immunosorbent Assay , Genotype , HIV Antibodies/blood , HIV Antibodies/genetics , HIV Infections/genetics , HIV-1/genetics , Humans , Hybridomas/immunology , Viral Load
14.
J Biosci Bioeng ; 113(2): 146-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079377

ABSTRACT

We generated genetically manipulated chickens and quail by infecting them with a retroviral vector expressing the human growth hormone under the control of chicken ovalbumin promoter/enhancer up to -3861 bp from the transcriptional start site. The growth hormone was expressed in an oviduct-specific manner and was found in egg white, although its level was low. The DNA sequence of the integrated form of the viral vector in the packaging cells was shown to be truncated and contained only the sequence spanning -3861 to -1569 bp. This represented only the DNase I hypersensitive site (DHS) III of the 4 DHSs and lacked the proximal promoter of the ovalbumin control region. We found several TATA-like and other promoter motifs of approximately -1800 bp and considered that these promoter motifs and DHS III may cause weak but oviduct-specific expression of the growth hormone. To prove this hypothesis and apply this system to oviduct-specific expression of the transgene, the truncated regulatory sequence was fused to an artificial transactivator-promoter system. In this system, initial weak but oviduct-specific expression of the Tet activator from the promoter element in the ovalbumin control sequence triggered a self-amplifying cycle of expression. DsRed was specifically expressed in oviduct cells of genetically manipulated chickens using this system. Furthermore, deletion of a short region possibly containing the promoter elements (-2112 to -1569 bp) completely abrogated oviduct-specific expression. Taken together, these results suggest that weak expression of this putative promoter causes oviduct-specific expression of the transgene.


Subject(s)
Chickens/genetics , Enhancer Elements, Genetic , Ovalbumin/genetics , Oviducts/metabolism , Transgenes , Animals , Animals, Genetically Modified , Chick Embryo , Chickens/metabolism , Female , Human Growth Hormone/genetics , Humans , Ovalbumin/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
15.
J Vet Med Sci ; 72(7): 893-901, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20215716

ABSTRACT

Dogs can be divided into two genetic groups (a minor HK phenotype and a major LK phenotype) based on erythrocyte monovalent cation concentrations, which are controlled by the putative hk and lk allelic genes. HK dogs retain Na,K-ATPase in their erythrocytes due to the high activity of the enzyme in their precursor cells, whereas total loss of reticulocyte Na,K-ATPase occurs in LK dogs. Here, we report that the levels of the lipid raft-associated membrane protein stomatin decrease in parallel with those of Na,K-ATPase during reticulocyte maturation due to its extrusion in exosomes. The stomatin content of HK reticulocytes is higher than that of LK reticulocytes, and remains in the erythrocytes at levels compatible with that in human erythrocytes. However, it is almost absent from LK erythrocytes with the lk/lk genotype; similar to the deficiency seen in human red cells with overhydrated stomatocytosis. LK erythrocytes from hk/lk genotype dogs show reduced, but not negligible, levels of stomatin. These results indicate that the erythrocyte stomatin level is a suitable genotypic marker for the HK/LK red cell phenotype, and suggests a functional association between stomatin and Na,K-ATPase. The absence of morphological abnormalities in the erythrocytes of stomatin-deficient LK dogs also confirms that stomatin deficiency and stomatocytic shape change are independent from each other.


Subject(s)
Dogs/genetics , Potassium/blood , Amino Acid Sequence , Anemia/blood , Anemia/veterinary , Animals , Antibodies, Monoclonal , Cations/blood , Dogs/blood , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/ultrastructure , Erythrocytes/metabolism , Genotype , Humans , Membrane Proteins/blood , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Pedigree , Phenotype , Reticulocytes/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sodium-Potassium-Exchanging ATPase/metabolism
16.
J Gastroenterol ; 45(1): 52-9, 2010.
Article in English | MEDLINE | ID: mdl-19789838

ABSTRACT

BACKGROUND AND AIMS: Serum biomarkers for the early detection of pancreatic cancer are not currently available. We evaluated the usefulness of a novel serum marker, REG4, in the diagnosis of pancreatic cancer, as compared to carbohydrate antigen (CA) 19-9. METHODS: We collected pretherapeutic sera from 92 patients with pancreatic cancer, as well as sera from 28 patients with other pancreatic tumors, 11 patients with pancreatitis, and 69 healthy controls. Serum levels of REG4 were measured using a standard sandwich enzyme-linked immunosorbent assay (ELISA). RESULTS: Compared with healthy controls, serum levels of REG4 were higher in pancreatic cancer patients (P < 0.001), and in patients with pancreatitis (P < 0.001). Receiver operating characteristic (ROC) analysis indicated that serum REG4 performed better than serum CA19-9 for distinguishing patients with pancreatic cancer from healthy controls [areas under the curve (AUC) for REG4 and CA19-9 were 0.922 and 0.884, respectively]. When we validated the study, the sensitivity of REG4 for pancreatic cancer was 94.9%, specificity was 64.0%, and accuracy was 77.5% for the REG4 cutoff value of 3.49 ng/ml. No correlation was seen between serum REG4 and CA19-9 levels, with the sensitivity, specificity, and accuracy of the combined markers reaching 100.0, 60.0, and 77.5%, respectively. No significant differences were seen among any stages of pancreatic cancer. In surgical specimens, immunohistochemical analysis found a correlation between serum REG4 levels and REG4 expression in pancreatic cancers. CONCLUSIONS: REG4 is expressed in pancreatic cancer, and serum levels of REG4 offer a useful indicator for distinguishing between patients with pancreatic cancer and healthy subjects. Serum REG4 has potential for use as a screening serum marker for pancreatic cancers, including early-stage cancers.


Subject(s)
Adenocarcinoma/diagnosis , Carcinoma, Pancreatic Ductal/diagnosis , Lectins, C-Type/blood , Pancreatic Neoplasms/diagnosis , Adenocarcinoma/pathology , Area Under Curve , Biomarkers, Tumor/blood , CA-19-9 Antigen/blood , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/pathology , Pancreatitis/pathology , Pancreatitis-Associated Proteins , ROC Curve , Sensitivity and Specificity
17.
J Biotechnol ; 141(1-2): 18-25, 2009 Apr 20.
Article in English | MEDLINE | ID: mdl-19428726

ABSTRACT

Genetically manipulated chickens producing chimeric monoclonal antibodies were generated by injecting retroviral vectors encoding genes for the heavy and light chains of antibodies into developing embryos. The transgene was detected in all chickens that hatched, and they stably produced the chimeric antibodies in their serum. After sexual maturation, the antibodies were also produced in eggs laid by the manipulated hens. The stable antibody production was observed both in egg white and yolk throughout the breeding period. The chimeric antibodies produced by the chickens were properly assembled and exhibited antigen-binding activities. Furthermore, we characterized the structures of the N-linked oligosaccharide chains added to the Fc-region of the recombinant antibodies produced in the serum, egg white and yolk of the chickens.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Chickens/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Chick Embryo , Egg White/chemistry , Egg Yolk/chemistry , Genetic Vectors/genetics , Oligosaccharides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Retroviridae/genetics
18.
J Biosci Bioeng ; 102(4): 297-303, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17116575

ABSTRACT

The use of transgenic avian species as a transgenic bioreactor for the production of recombinant proteins has been proposed. In recent years, although various procedures for generating transgenic chickens have been reported, the expression of a useful protein at a commercially feasible level has rarely been attained. In this study, we injected a concentrated retroviral vector into quail embryos to generate genetically manipulated quails that produce recombinant proteins. We found that transgene expression in the whole body at a high level was observed for viral injection into the heart of the developing embryos after a 48-h incubation. For the practical production of a useful protein, a retroviral vector encoding an anti-prion scFv-Fc gene under the control of the beta-actin promoter was injected into quail embryos. The quails that hatched stably produced scFv-Fc at a high level in their serum and egg white. The production of scFv-Fc was maintained throughout the breeding period. scFv-Fc purified from the egg white retained the antigen-binding activity. This system exhibited the potential of transgenic quails for the commercial production of recombinant proteins.


Subject(s)
Coturnix/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Variable Region/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Transfection/methods , Animals , Animals, Genetically Modified , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Variable Region/genetics , Promoter Regions, Genetic/genetics
19.
J Cell Sci ; 119(Pt 17): 3602-12, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16912075

ABSTRACT

Various mutations in the AE1 (anion exchanger 1, band 3) gene cause dominant hereditary spherocytosis, a common congenital hemolytic anemia associated with deficiencies of AE1 of different degrees and loss of mutant protein from red blood cell membranes. To determine the mechanisms underlying decreases in AE1 protein levels, we employed K562 and HEK293 cell lines and Xenopus oocytes together with bovine wild-type AE1 and an R664X nonsense mutant responsible for dominant hereditary spherocytosis to analyze protein expression, turnover, and intracellular localization. R664X-mutant protein underwent rapid degradation and caused specifically increased turnover and impaired trafficking to the plasma membrane of the wild-type protein through hetero-oligomer formation in K562 cells. Consistent with those observations, co-expression of mutant and wild-type AE1 reduced anion transport by the wild-type protein in oocytes. Transfection studies in K562 and HEK293 cells revealed that the major pathway mediating degradation of both R664X and wild-type AE1 employed endoplasmic reticulum (ER)-associated degradation through the proteasomal pathway. Proteasomal degradation of R664X protein appeared to be independent of both ubiquitylation and N-glycosylation, and aggresome formation was not observed following proteasome inhibition. These findings indicate that AE1 R664X protein, which is associated with dominant hereditary spherocytosis, has a dominant-negative effect on the expression of wild-type AE1.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Endoplasmic Reticulum/metabolism , Spherocytosis, Hereditary/metabolism , Ubiquitins/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Cattle , Cell Line , Cell Membrane/metabolism , Genes, Dominant , Humans , Mutation , Proteasome Endopeptidase Complex/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spherocytosis, Hereditary/genetics
20.
J Biosci Bioeng ; 102(6): 518-23, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17270716

ABSTRACT

We examined the transport of human immunoglobulin G (IgG) subclasses and fusion proteins with the Fc region of human IgG to the egg yolk, after the proteins were injected into a vein of hens. Human IgGs were efficiently transported and accumulated into the yolk, whereas the proteins were not detected in the egg white. Among human IgG subclasses, IgG2 was transported most efficiently. Fc-fusion proteins injected were also transported into the yolk. A fusion protein with the Fc region derived from human IgG2 was more efficiently transported into the yolk than the counterpart fusion with the Fc region from human IgG1. This study shows that the recovery of recombinant antibodies and Fc-fusion proteins from the yolk is an effective method in transgenic chicken bioreactors.


Subject(s)
Egg Yolk/metabolism , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Animals , Biological Transport, Active/physiology , Chick Embryo , Female , Humans , Maternal-Fetal Exchange , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...