ABSTRACT
Recombinant human anti-tumor necrosis factor (TNF)-α scFv-Fc was expressed in TKO mutant Arabidopsis thaliana seeds using plant-specific codons. Immunoblotting using a human IgG1 antibody detected the expression of anti-TNF-α proteins in plants. Results from qRT-PCR analysis demonstrated that the time of harvest significantly affected the protein yield and quality. Our results indicate that the Phaseolus vulgaris ß-phaseolin promoter directed anti-TNF-α scFv-Fc expression in A. thaliana seeds, with a maximum yield obtained at 20-days of development. Although the yield of anti-TNF-α scFv-Fc protein was not very high, accumulation of recombinant proteins in seeds is an attractive and simple method that can be used to purify biologically active anti-TNF-α scFv-Fc.
Subject(s)
Arabidopsis/genetics , Immunoglobulin Fc Fragments/genetics , Seeds/metabolism , Single-Chain Antibodies/genetics , Transgenes , Tumor Necrosis Factor-alpha/immunology , Arabidopsis/metabolism , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Recombinant Proteins , Seeds/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolismABSTRACT
To identify the linear epitope for Fc-binding to the mouse immunoglobulin G (IgG) Fc receptor (moFcγRI), peptides derived from the membrane-distal extracellular domain (EC2) of moFcγRI, corresponding to the homologous region of human FcγRI (huFcγRI) and huFcγRII, were synthesized. Using a dot-blot assay, six peptides were tested. The results showed that the moRI3 peptide (CVFYRNGKSFQFS) could combine with mouse IgG efficiently. A competitive enzyme-linked immunosorbent assay (ELISA) showed that the IC50 value of the moRI3 peptide was 38.03 mM. The moRI3 peptide could inhibit the combination of mouse IgG to the transfected COS 7 cells significantly with an IC50 value of 72.68 mM. The IgG-binding region of moFcγRI was also localized in the C'-E loop of the EC2 domain as predicted according to huFcγRI and huFcγRII. We predicted that the minimum effective IgG-binding region of moFcγRI may be the peptide 153SFQFSS158. The linear epitope for immunoglobulin-binding to mouse FcγR is also described. Thus, we generated a peptide that targets a fundamental aspect of ligand recognition by this receptor class.
Subject(s)
Epitopes/chemistry , Immunoglobulin Fc Fragments/metabolism , Peptide Fragments/chemical synthesis , Receptors, IgG/metabolism , Animals , COS Cells , Chlorocebus aethiops , Epitope Mapping , Epitopes/immunology , Epitopes/metabolism , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/metabolism , Mice , Peptide Fragments/immunology , Peptide Fragments/metabolismABSTRACT
Excellent results regarding improved therapeutic properties have been often obtained through the conversion of a single-chain variable fragment (scFv) into a noncovalent dimeric antibody (diabody) via peptide linker shortening. We utilized this approach to obtain a dimeric version of the human scFv 6009F, which was originally engineered to neutralize the Cn2 toxin of Centruroides noxius scorpion venom. However, some envenoming symptoms remained with diabody 6009F. Diabody 6009F was subjected to directed evolution to obtain a variant capable of eliminating envenoming symptoms. After two rounds of biopanning, diabody D4 was isolated. It exhibited a single mutation (E43G) in framework 2 of the heavy-chain variable domain. Diabody D4 displayed an increase in T(m) (thermal transition midpoint temperature) of 6.3°C compared with its dimeric precursor. The importance of the E43G mutation was tested in the context of the human scFv LR, a highly efficient antibody against Cn2, which was previously generated by our group [Riaño-Umbarila, L., Contreras-Ferrat, G., Olamendi-Portugal, T., Morelos-Juárez, C., Corzo, G., Possani, L. D. and Becerril, B. (2011). J. Biol. Chem.286, 6143-6151]. The new variant, scFv LER, displayed an increase in T(m) of 3.4°C and was capable of neutralizing 2 LD(50) of Cn2 toxin with no detectable symptoms when injected into mice at a 1:1 toxin-to-antibody molar ratio. These results showed that the E43G mutation might increase the therapeutic properties of these antibody fragments. Molecular modeling and dynamics results suggest that the rearrangement of the hydrogen-bonding network near the E43G mutation could explain the improved functional stability and neutralization properties of both the diabody D4 and scFv LER.
Subject(s)
Immunoglobulin Fc Fragments , Immunoglobulin Heavy Chains , Scorpion Venoms/immunology , Single-Chain Antibodies , Amino Acid Sequence , Animals , Antibodies, Neutralizing , Antibody Affinity , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mice , Molecular Dynamics Simulation , Neutralization Tests , Protein Binding/immunology , Protein Structure, Tertiary , Scorpion Venoms/chemistry , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Surface Plasmon ResonanceABSTRACT
Ischemia reperfusion injury is characterized by local and systemic inflammation leading to considerable mortality. Previously, we have reported that soluble T1/ST2 (sST2), a member of the IL-1 receptor gene family, inhibits LPS-induced macrophage proinflammatory cytokine production. Here, we report the therapeutic effect of sST2-Fc in a murine model of intestinal ischemia reperfusion-induced injury. Administration of sST2-Fc fusion protein i.v., 10 min before reperfusion, reduced the production of TNF-alpha dose-dependently in the intestine and in the lungs. The sST2-Fc treatment with the highest dose (100 mug) resulted in inhibited vascular permeability, neutrophilia, and hemorrhage in the intestine and the lungs compared with controls treated with normal IgG. This was associated with down-regulated tissue levels of proinflammatory cytokines, markedly reduced serum TNF-alpha levels, and increased survival of mice from the sST2-Fc-treated group after ischemia and reperfusion injury. The beneficial effect of sST2-Fc treatment was associated with elevated IL-10 production in intestine and lung. sST2-Fc was not able to prevent the inflammatory response associated with intestinal ischemia and reperfusion in IL-10-deficient mice, suggesting that sST2 exerts its anti-inflammatory effect in a IL-10-dependent manner. These results also demonstrate that sST2-Fc may provide a novel, complementary approach in treating ischemic reperfusion injury.
Subject(s)
Immunoglobulin Fc Fragments/therapeutic use , Intestines/pathology , Membrane Proteins/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Animals , Disease Models, Animal , Dose-Response Relationship, Immunologic , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Inflammation , Interleukin-1 Receptor-Like 1 Protein , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Intestines/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Reperfusion Injury/immunology , Survival Rate , Time Factors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesisABSTRACT
Monocytes (Mo) mediate central functions in inflammation and immunity. Different subpopulations of Mo with distinct phenotype and functional properties have been described. Here, we investigate the phenotype and function of peripheral Mo from children with hemolytic uremic syndrome (HUS). For this purpose, blood samples from patients in the acute period of HUS (HUS AP) were obtained on admission before dialysis and/or transfusion. The Mo phenotypic characterization was performed on whole blood by flow cytometry, and markers associated to biological functions were selected: CD14 accounting for lipopolysaccharide (LPS) responsiveness, CD11b for adhesion, Fc receptor for immunoglobulin G type I (FcgammaRI)/CD64 for phagocytosis and cytotoxicity, and human leukocyte antigen (HLA)-DR for antigen presentation. Some of these functions were also determined. Moreover, the percentage of CD14+ CD16+ Mo was evaluated. We found that the entire HUS AP Mo population exhibited reduced CD14, CD64, and CD11b expression and decreased LPS-induced tumor necrosis factor production and Fcgamma-dependent cytotoxicity. HUS AP showed an increased percentage of CD14+ CD16+ Mo with higher CD16 and lower CD14 levels compared with the same subset from healthy children. Moreover, the CD14++ CD16- Mo subpopulation of HUS AP had a decreased HLA-DR expression, which correlated with severity. In conclusion, the Mo population from HUS AP patients presents phenotypic and functional alterations. The contribution to the pathogenesis and the possible scenarios that led to these changes are discussed.
Subject(s)
Antigens, Differentiation/immunology , Fetal Blood/immunology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/physiopathology , Monocytes/immunology , Cell Count , Child, Preschool , Cytotoxicity, Immunologic , Flow Cytometry , Hemolytic-Uremic Syndrome/diagnosis , Humans , Immunoglobulin Fc Fragments/immunology , Infant , Lipopolysaccharides/pharmacology , Phenotype , Tumor Necrosis Factors/immunologySubject(s)
Immunoglobulin Fc Fragments/immunology , Staphylococcal Protein A/immunology , Animals , Electrophoresis, Capillary/methods , Immunoglobulin Fc Fragments/isolation & purification , Immunoglobulin G/immunology , Kinetics , Mice , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Potentiometry/methods , Recombinant Fusion Proteins/immunology , Staphylococcal Protein A/chemistryABSTRACT
The IL-1 receptor-related protein T1 is expressed on the surface of Th2, but not Th1 cells. Studies with anti-T1 monoclonal antibodies have suggested that T1 is critical for development of normal Th2-type responses. To elucidate the role of T1 in vivo, we generated T1-deficient mice and a T1-transgenic strain which secretes soluble T1-Fc fusion protein into the serum. These were analyzed for the Th2 immune response induced by infection with the parasitic nematode Nippostrongylus brasiliensis. Although Th2 cytokine production by lymph node cells was similar in all groups of N. brasiliensis-infected mice, a decrease in IL-5 production by lung lymphocytes was detected in both T1-deficient and T1-Fc-transgenic mice compared to control littermates. This difference in IL-5 production did not influence blood eosinophilia, but recruitment of eosinophils into lung tissue, especially in T1-Fc-transgenic mice was slightly decreased. However, induction of all other immune parameters was normal and both T1-deficient and T1-Fc-transgenic mice were able to clear the parasite infection within 12 days with kinetics similar to those in control mice. Therefore, in contrast to previous suggestions, we conclude that the T1 protein is not obligatory for normal development of Th2 immune responses.
Subject(s)
Immunoglobulin gamma-Chains/immunology , Membrane Proteins , Nippostrongylus/immunology , Proteins/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Eosinophilia/immunology , Female , Gene Expression , Humans , Immunoglobulin E/blood , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/blood , Immunoglobulin gamma-Chains/genetics , Interleukin-1 Receptor-Like 1 Protein , Interleukin-5/biosynthesis , Kinetics , Lung/cytology , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proteins/genetics , Rats , Rats, Inbred Lew , Receptors, Cell Surface , Receptors, Interleukin , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunologyABSTRACT
Passive transfer of immune serum obtained from mice chronically infected with Trypanosoma cruzi to mice containing circulating bloodstream trypomastigotes induces a very fast clearance of the parasites. Comparison of trypomastigotes clearance in normocomplementemic and C5-deficient mice showed no difference. IgG fraction obtained from immune serum was very efficient at inducing complement-mediated lysis and immune clearance of bloodstream trypomastigotes, whereas its Fc-missing F (ab') 2 fragments, although able to induce lysis, were unable to induce clearance. It is suggested that the immune clearance of bloodstream trypomastigotes is dependent on the antibody Fc region and that complement-mediated lysis is not a prerequisite for elimination of the parasites from circulation.
Subject(s)
Chagas Disease/immunology , Immunization, Secondary , Immunoglobulin Fc Fragments/immunology , Animals , Antibodies, Protozoan/immunology , Chagas Disease/therapy , Complement C5/deficiency , Female , Immunoglobulin Fab Fragments/immunology , Male , Mice , Mice, Inbred StrainsABSTRACT
We have previously characterized with monoclonal antibodies (MAbs) seven unique epitopes on the two envelope glycoproteins of Venezuelan equine encephalomyelitis (VEE) virus vaccine strain TC-83. The epitopes important in protection from VEE virus infection were determined in passive antibody transfer studies, with virulent VEE (Trinidad donkey) virus as the challenge virus. Selected high-avidity MAbs to the three major protective epitopes (E2c, E1b, and E1d) were assayed for in vitro complement activity. All three fixed murine complement to high titer. Limited pepsin digestion of the anti-E2c in the presence of cysteine resulted in a rapid decrease and complete loss of complement-fixing ability by 2 h, but the majority of mice, except at the lowest dilution of MAb, were protected until the Fc termini were cleaved at 3 h. Anti-E2c F(ab')2 would neutralize VEE (Trinidad donkey) virus more efficiently than either Fab' or Fab; none of the fragments would fix complement or was effective in passive protection. C5-deficient mice and mice depleted of C3 with cobra venom factor were still protected from VEE (Trinidad donkey) virus challenge after passive transfer of either anti-E2c or anti-E1b MAb. The results show that the anti-E2c MAb mediates neutralization through bivalent binding at a critical site on the virion and that Fc effector functions, other than complement, are necessary for protection. Although the ability of the anti-E2c MAb to fix complement was associated with its ability to protect in vivo, no direct cause-and-effect relationship was found. Since the epitope defined by the anti-E1d antibody is found on the cell membrane, but is not expressed on the infectious virion, protection in mice was most likely mediated at the cellular level, possibly by inhibition of the final stages of virion maturation.
Subject(s)
Complement System Proteins/immunology , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fragments/immunology , Animals , Antibodies, Monoclonal , Epitopes , Glycoproteins/immunology , Mice , Neutralization Tests , Pepsin A/metabolismABSTRACT
We have investigated the nature of the ligand involved in antibody-dependent cellular cytotoxicity (ADCC) of human leukocytes to epimastigotes of Trypanosoma cruzi. Purified anti-T. cruzi IgG was highly efficient in mediating ADCC whereas IgM mediated the killing of this parasite poorly even at high concentrations. The presence of the Fc portion on the IgG molecule seems to be necessary since F(ab')2 derived from anti-T. cruzi IgG did not mediate ADCC. We also present evidence suggesting that the mediator, aside from promoting the interaction between the effector and target cells, may play a functional role in triggering target destructions by the effector cells. This conclusion is based on results of experiments in which lectins capable of binding to both leukocytes and parasite were used as mediators of cellular cytotoxicity. The lectin concanavalin A could readily replace IgG for human leukocyte killing of T. cruzi. In contrast, lectins from Lens culinaris, Triticum vulgaris, Aaptos papillata and Bandeirae simplicifolia although capable of interacting with leukocytes and T. cruzi, did not mediate the cellular cytotoxicity of the parasites. The specific cellular mechanism of parasite killing, i.e. phagocytosis and or extracellular lysis, remains to be determined.