Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Publication year range
1.
Chinese Journal of Biologicals ; (12): 21-25+31, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-965573

ABSTRACT

@#Abstract:Objective To improve the replication level of varicella⁃zoster virus(VZV)in human diploid cell line MRC⁃5 and increase the yield of VZV vaccine by reducing the expression of interferon(IFN)related genes via optimizing the cell line MRC⁃5. Methods Interferon receptor 1(IFNAR1)silenced MRC⁃5 cell line(MRC⁃5IFNAR1⁃)was constructed by CRISPR/Cas9 gene editing technology,which was determined for the relative expression of IFNAR1 mRNA,and for those of mRNA of IFN related genes IFNβ and OAS1 after VZV infection by qRT⁃PCR to evaluate the effect of gene silencing. Gene mutation sequences were further identified by sequencing of the silenced sites. The replication of VZV in MRC⁃5 and MRC⁃5IFNAR1⁃ cell lines was compared 168 h after VZV infection by using qRT⁃PCR and plaque formation unit(PFU)assay, to evaluate the effect of MRC⁃5IFNAR1⁃cell line on VZV replication. Results The growth status of MRC⁃5IFNAR1⁃ cell line wasconsistent with that of MRC ⁃ 5 cells,and the relative expression of IFNAR1 mRNA decreased by 73%;The relative expressions of IFNβ and OAS1 mRNA in MRC⁃5IFNAR1⁃ cell line were 61% and 90% lower than those in MRC⁃ 5 cells respectively after VZV infection;In addition,168 h after VZV infection,the level of DNA replication and the titer of VZV increased by 5. 7 folds and 4 folds respectively. Conclusion The successful establishment of MRC⁃5IFNAR1⁃ cell line may be a potential scheme to increase the yield of vaccines based on human diploid cells,and provided a reference for expanding production of VZV vaccine.

2.
PLoS One ; 8(8): e71383, 2013.
Article in English | MEDLINE | ID: mdl-23977032

ABSTRACT

Post-exposure prophylactic (PEP) neutralizing antibodies against Rabies are the most effective way to prevent infection-related fatality. The outer envelope glycoprotein of the Rabies virus (RABV) is the most significant surface antigen for generating virus-neutralizing antibodies. The small size and uncompromised functional specificity of single domain antibodies (sdAbs) can be exploited in the fields of experimental therapeutic applications for infectious diseases through formatting flexibilities to increase their avidity towards target antigens. In this study, we used phage display technique to select and identify sdAbs that were specific for the RABV glycoprotein from a naïve llama-derived antibody library. To increase their neutralizing potencies, the sdAbs were fused with a coiled-coil peptide derived from the human cartilage oligomeric matrix protein (COMP48) to form homogenous pentavalent multimers, known as combodies. Compared to monovalent sdAbs, the combodies, namely 26424 and 26434, exhibited high avidity and were able to neutralize 85-fold higher input of RABV (CVS-11 strain) pseudotypes in vitro, as a result of multimerization, while retaining their specificities for target antigen. 26424 and 26434 were capable of neutralizing CVS-11 pseudotypes in vitro by 90-95% as compared to human rabies immunoglobulin (HRIG), currently used for PEP in Rabies. The multimeric sdAbs were also demonstrated to be partially protective for mice that were infected with lethal doses of rabies virus in vivo. The results demonstrate that the combodies could be valuable tools in understanding viral mechanisms, diagnosis and possible anti-viral candidate for RABV infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/prevention & control , Single-Domain Antibodies/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/classification , Antibodies, Neutralizing/genetics , Antibodies, Viral/classification , Antibodies, Viral/genetics , Camelids, New World , Cartilage Oligomeric Matrix Protein/genetics , Cartilage Oligomeric Matrix Protein/immunology , Escherichia coli/genetics , Humans , Mice , Peptide Library , Phylogeny , Protein Multimerization , Rabies/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Domain Antibodies/classification , Single-Domain Antibodies/genetics , Viral Envelope Proteins/genetics
3.
Int Immunopharmacol ; 17(1): 9-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23624214

ABSTRACT

As an ideal tumor antigen, survivin has been widely used for tumor immunotherapy. Nevertheless, no effective protein vaccine targeting survivin has been reported, which may be due to its poor ability to induce cellular immunity. Thus, a suitable immunoadjuvant and optimized immunization strategy can greatly enhance the cellular immune response to this protein vaccine. DDA/MPL (monophosphoryl lipid A formulated with cationic dimethyldioctadecylammonium) has been reported to enhance the antigen uptake and presentation to T cells as an adjuvant. Meanwhile, a heterologous prime-boost strategy can enhance the cellular immunity of a protein vaccine by applying different antigen-presenting systems. Here, DDA/MPL and an adenovirus prime-protein boost strategy were applied to enhance the specific anti-tumor immunity of a truncated survivin protein vaccine. Antigen-specific IFN-γ-secreting T cells were increased by 10-fold, and cytotoxic T lympohocytes (CTLs) were induced effectively when the protein vaccine was combined with the DDA/MPL adjuvant. Meanwhile, the Th1 type cellular immune response was strongly enhanced and tumor inhibition was significantly increased by 96% with the adenovirus/protein prime-boost strategy, compared to the protein homologous prime-boost strategy. Moreover, this adjuvanted heterologous prime-boost strategy combined with oxaliplatin could significantly enhance the efficiency of tumor growth inhibition through promoting the proliferation of splenocytes. Thus, our results provide a novel vaccine strategy for cancer therapy using an adenovirus prime-protein boost strategy in a murine melanoma model, and its combination with oxaliplatin may further enhance the anti-tumor efficacy while alleviating side effects of the drug.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Lipid A/analogs & derivatives , Melanoma/therapy , Quaternary Ammonium Compounds/pharmacology , Adjuvants, Immunologic , Animals , Cancer Vaccines , Cell Line , Cell Proliferation , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Lipid A/administration & dosage , Lipid A/pharmacology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , Organoplatinum Compounds , Oxaliplatin , Quaternary Ammonium Compounds/administration & dosage , Specific Pathogen-Free Organisms , Survivin , T-Lymphocyte Subsets
4.
Amino Acids ; 43(6): 2431-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22674377

ABSTRACT

The deuterohemin-peptide conjugate, DhHP-6 (Dh-ß-AHTVEK-NH(2)), is a microperoxidase mimetic, which has demonstrated substantial benefits in vivo as a scavenger of reactive oxygen species (ROS). In this study, specific multi-site N-methylated derivatives of DhHP-6 were designed and synthesized to improve metabolic stability and intestinal absorption, which are important factors for oral delivery of therapeutic peptides and proteins. The DhHP-6 derivatives were tested for (1) scavenging potential of hydrogen peroxide (H(2)O(2)); (2) permeability across Caco-2 cell monolayers and everted gut sacs; and (3) enzymatic stability in serum and intestinal homogenate. The results indicated that the activities of the DhHP-6 derivatives were not influenced by N-methylation, and that tri-N-methylation of DhHP-6 could significantly increase intestinal flux, resulting in a two- to threefold higher apparent permeability coefficient. In addition, molecules with N-methylation at selected sites (e.g., Glu residue) showed high resistance against proteolytic degradation in both diluted serum and intestinal preparation, with 50- to 140-fold higher half-life values. These findings suggest that the DhHP-6 derivatives with appropriate N-methylation could retain activity levels equivalent to that of the parent peptide, while showing enhanced intestinal permeability and stability against enzymatic degradation. The tri-N-methylated peptide Dh-ß-AH(Me)T(Me)V(Me)EK-NH(2) derived from this study may be developed as a promising candidate for oral administration.


Subject(s)
Hemin/analogs & derivatives , Intestinal Mucosa/metabolism , Oligopeptides/metabolism , Peroxidase/metabolism , Animals , Caco-2 Cells , Enzyme Stability , Hemin/chemical synthesis , Hemin/chemistry , Hemin/metabolism , Humans , Intestinal Mucosa/chemistry , Methylation , Mice , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Permeability , Peroxidase/chemistry , Substrate Specificity
5.
Mol Immunol ; 51(2): 188-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484084

ABSTRACT

Rabies is a fatal infectious disease requiring efficient protection provided by post-exposure prophylaxis (PEP) with rabies immunoglobulin (RIG). The single-chain Fv fragment (scFv) is a small engineered antigen binding protein derived from antibody variable heavy (V(H)) and light (V(L)) chains. This novel antibody format may potentially replace the current application of RIG to detect and neutralize rabies virus (RV). However, the broad use of scFvs is confined by their generally low stability. In this study, a scFv (FV57) was constructed based on the monoclonal antibody, MAB57, against RV. To enhance its stability and neutralizing potency, a disulfide-stabilized scFv, ds-FV57, was also derived by introduction of cysteines at V(H)44 and V(L)100. Furthermore, the cysteine at V(L)85 of ds-FV57 was mutated to serine to construct ds-FV57(VL85Ser) in order to avoid potential mis-formed disulfide bonds which would alter the affinity of the scFv. The stability and activity of all three proteins expressed in Escherichia coli were evaluated. All of the constructed scFvs could provide efficient protection against RV infection both in vivo and in vitro. However, the stability of ds-FV57(VL85Ser) was notably improved, and its in vitro neutralizing potency against RV infection was enhanced. Our findings from these stabilization modifications support the feasibility of developing scFvs for PEP treatment of rabies.


Subject(s)
Post-Exposure Prophylaxis/methods , Rabies Vaccines/pharmacology , Rabies/prevention & control , Single-Chain Antibodies/pharmacology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibody Affinity , Cricetinae , Mice , Protein Binding , Protein Engineering/methods , Protein Stability , Rabies Vaccines/chemistry , Rabies Vaccines/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology
6.
Protein Pept Lett ; 18(11): 1099-106, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21675946

ABSTRACT

Single-chain Fv fragment (scFv) of anti-rabies glycoprotein (G protein) has been recommended as a new agent for detecting and neutralizing lethal rabies virus. In this study, we constructed scFv that corresponded to the FV fragment of CR57, a monoclonal antibody against rabies virus, and called it FV57. Despite its virus neutralization activity, FV57 may or may not recognize the same epitope as that recognized by CR57. To resolve this issue, the binding epitope of rabies virus G protein recognized by FV57 was identified. A recombinant rabies virus G protein fragment (RVG179; residues 179-281) comprising several epitopes was expressed in E.coli, purified, and the specificity of its binding with FV57 was determined. In addition, a peptide (abbreviated as EP, residues 224-236) comprising the known epitope of G protein to which CR57 binds was synthesized and the potency of its binding with FV57 was also determined. The results showed that FV57 could specifically bind to RVG179 and EP. Competitive ELISA experiments indicated that RVG179 and EP were able to compete with the rabies virus G protein for binding with FV57. Since no other epitope within residues 224- 236 has been reported, except for the epitope to which CR57 binds (residues 226-231), the epitope recognized by FV57 was the same as its intact antibody CR57. This demonstrated that the complementarity-determining regions (CDRs) of the heavy and light chains of FV57 have folded into the correct conformation as those of CR57.


Subject(s)
Antigens, Viral/chemistry , Antigens, Viral/immunology , Epitope Mapping/methods , Glycoproteins/chemistry , Glycoproteins/immunology , Rabies virus/immunology , Single-Chain Antibodies/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Line , Cricetinae , Enzyme-Linked Immunosorbent Assay , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...